US20200002727A1 - Multi-site specific integration cells for difficult to express proteins - Google Patents

Multi-site specific integration cells for difficult to express proteins Download PDF

Info

Publication number
US20200002727A1
US20200002727A1 US16/486,363 US201816486363A US2020002727A1 US 20200002727 A1 US20200002727 A1 US 20200002727A1 US 201816486363 A US201816486363 A US 201816486363A US 2020002727 A1 US2020002727 A1 US 2020002727A1
Authority
US
United States
Prior art keywords
gene
cell
locus
interest
rts
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US16/486,363
Other languages
English (en)
Inventor
Marc Feary
Robert J. Young
Mark Moffat
Gerald Fries Casperson
Heather Laurence JONES
Lin Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lonza AG
Pfizer Inc
Original Assignee
Lonza AG
Pfizer Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lonza AG, Pfizer Inc filed Critical Lonza AG
Priority to US16/486,363 priority Critical patent/US20200002727A1/en
Publication of US20200002727A1 publication Critical patent/US20200002727A1/en
Assigned to LONZA LTD reassignment LONZA LTD ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Feary, Marc, YOUNG, ROBERT J.
Assigned to PFIZER INC. reassignment PFIZER INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CASPERSON, GERALD FRIES, JONES, HEATHER LAURENCE, MOFFAT, MARK ALLEN, ZHANG, LIN
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3007Carcino-embryonic Antigens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/30Vector systems comprising sequences for excision in presence of a recombinase, e.g. loxP or FRT

Definitions

  • the present disclosure relates to a site-specific integration (SSI) mammalian cell that comprises at least two distinct recombination target sites (RTS) wherein two RTS are chromosomally-integrated within the NL1 locus or the NL2 locus.
  • the disclosure also relates to a SSI mammalian cell comprising at least four distinct RTS wherein two RTS are chromosomally-integrated within the NL1 or the NL2 locus and two RTS are chromosomally-integrated within a separate locus.
  • the disclosure also relates to methods for using the SSI mammalian host cell line to produce recombinant protein expression cell lines that can additionally express difficult to express proteins.
  • Biopharmaceuticals both novel and biosimilar, continue to see high demand and increasing sales revenues.
  • mAb non-monoclonal antibody
  • rP recombinant proteins
  • DtE difficult to express
  • DtE proteins can be associated with lower than expected titres or problems with expression of multiple chains, ancillary proteins, product recovery or purification (Pybus et al. Biotechnol. Bioeng. 111:372-85 (2014)).
  • DtE proteins include but are not limited to Fc-fusion proteins, bi- and tri-specific MAbs, enzymes, membrane receptors, and bi-specific T-cell engager BITE® (Micromet AG, Kunststoff, Germany) molecules and selected mAbs.
  • SSI site-specific integration
  • “Landing pads” located in the genomes of SSI cell lines can utilize recombination target sites (RTS) derived from site-specific recombinase systems such as the Saccharomyces cerevisiae -derived FLP-Frt system or the bacteriophage P1 derived Cre-loxP system.
  • RTS recombination target sites
  • the recombination enzyme or recombinase is responsible for recombination events between donor and target DNA containing compatible recombination sites (Frt or loxP respectively) (see, e.g., Wirth et al. Curr. Op. in Biotech. 18:411-9 (2007)).
  • recombinase-mediated cassette-exchange The process of integrating cassettes in SSI cell lines is referred to as recombinase-mediated cassette-exchange (RMCE).
  • RMCE recombinase-mediated cassette-exchange
  • cell line construction for recombinant protein production using RMCE generally involves co-transfection of an expression vector encoding the recombinase along with the targeting expression vector, containing the gene of interest (encoding the rP) and a selection marker flanked by recombinase targeting sequences.
  • SSI-generated cell lines that use a single landing pad can also have limitations. For example, such an approach usually, by design, results in a low number of integrated gene copies that could indirectly limit rP production recombinant protein expression titres. If production of multiple proteins in an SSI host cell line containing a single landing pad is required for rP production, all of the required genes might need to be included into a single vector.
  • One method to increase integrated copies of recombinant genes is accumulative SSI (sometimes called stacking or multiplexing, see, e.g., Kameyama et al. Biotechnol. Bioeng. 105:1106-14 (2010), Kawabe et al. Cytotechnology 64:267-79 (2012) and Turan et al.
  • the present invention fulfills this need by using tandem SSI landing pads—where two or more landing pads are integrated at the same loci may overcome the limitations of cumulative site-specific integration. In such a system, the landing pads are independently addressable (due to recombination site and selection marker choice).
  • the present disclosure provides a mammalian cell comprising at least two distinct RTS wherein two RTS are chromosomally-integrated within the NL1 locus or the NL2 locus.
  • the cell comprises two distinct RTS.
  • the two distinct RTS are chromosomally-integrated within the NL1 locus.
  • the two distinct RTS are chromosomally-integrated within the NL2 locus.
  • the cell comprises four distinct RTS.
  • the four distinct RTS are chromosomally-integrated on the same locus.
  • two distinct RTS are chromosomally-integrated on a separate locus.
  • the separate locus is the Fer1L4 locus.
  • two distinct RTS are chromosomally-integrated within the NL1 locus, and two distinct RTS are chromosomally-integrated within the NL2 locus.
  • the cell comprises six distinct RTS.
  • at least four distinct RTS are chromosomally-integrated on the same locus.
  • at least two distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus, and at least two distinct RTS are chromosomally-integrated on a separate locus.
  • the separate locus is the Fer1L4 locus.
  • At least two distinct RTS are chromosomally-integrated within the NL1 locus, and at least two distinct RTS are chromosomally-integrated within the NL2 locus.
  • at least one of the RTS is an Frt site, a lox site, a rox site, or an att site.
  • at least one of the RTS is selected from among SEQ ID Nos.: 1-30.
  • the cell is a mouse cell, a human cell, a Chinese hamster ovary (CHO) cell, a CHO-K1 cell, a CHO-DXB11 cell, a CHO-DG44 cell, a CHOK1SVTM cell including all variants, a CHOK1SV GS-KOTM (glutamine synthetase knockout) cell including all variants, a HEK cell, a HEK293 cell including adherent and suspension-adapted variants, a HeLa cell, or a HT1080 cell.
  • the cell is a HEK cell.
  • the cell comprises a first gene of interest, wherein the first gene of interest is chromosomally-integrated.
  • the first gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the DtE protein is selected from the group consisting of Fc-fusion protein, an enzyme, a membrane receptor, a bi-specific T-cell engager (BITE® Micromet AG, Kunststoff, Germany), or a monoclonal antibody.
  • the monoclonal antibody is a bi-specific monoclonal antibody or a tri-specific monoclonal antibody.
  • the first gene of interest is located between two of the RTS. In some embodiments, the first gene of interest is located within the NL1 locus.
  • the cell comprises a second gene of interest, wherein the second gene of interest is chromosomally-integrated. In some embodiments, the second gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof. In some embodiments, the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the DtE protein is selected from the group consisting of a Fc-fusion protein, an enzyme, a membrane receptor, a bi-specific T-cell engager (BITE®), or a monoclonal antibody.
  • the second gene of interest is located between two of the RTS. In some embodiments, the second gene of interest is located within the NL1 locus or the NL2 locus. In some embodiments, the first gene of interest is located within the NL1 locus, and the second gene of interest is located within the NL2 locus. In some embodiments, the cell comprises a third gene of interest, wherein the third gene of interest is chromosomally-integrated.
  • the third gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the third gene of interest is located between two of the RTS.
  • the third gene of interest is located within the NL1 locus or the NL2 locus.
  • the third gene of interest is located within a locus distinct from the NL1 locus and the NL2 locus.
  • the first gene of interest, the second gene of interest, and the third gene of interest are within three separate loci.
  • the cell comprises a site-specific recombinase gene. In some embodiments, the site-specific recombinase gene is chromosomally-integrated.
  • the present disclosure provides a mammalian cell comprising at least four distinct RTS, wherein the cell comprises (a) at least two distinct RTS are chromosomally-integrated within the NL1 locus or NL2 locus; (b) a first gene of interest is integrated between the at least two RTS of (a), wherein the first gene of interest comprises a reporter gene, a gene encoding a DtE protein, an ancillary gene or a combination thereof; (c) and a second gene of interest is integrated within a second chromosomal locus distinct from the locus of (a), wherein the second gene of interest comprises a reporter gene, a gene encoding a DtE protein, an ancillary gene or a combination thereof.
  • the present disclosure provides a mammalian cell comprising at least four distinct RTS, wherein the cell comprises (a) at least two distinct RTS are chromosomally-integrated within the Fer1L4 locus; (b) at least two distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus; (c) a first gene of interest is chromosomally-integrated within the Fer1L4 locus, wherein the first gene of interest comprises a reporter gene, a gene encoding a DtE protein, an ancillary gene or a combination thereof; and (d) a second gene of interest is chromosomally-integrated within the within the NL1 locus or NL2 locus of (b), wherein the second gene of interest comprises a reporter gene, a gene encoding a DtE protein, an ancillary gene or a combination thereof.
  • the present disclosure provides a mammalian cell comprising at least six distinct RTS, wherein the cell comprises (a) at least two distinct RTS and a first gene of interest are chromosomally-integrated within the Fer1L4 locus; (b) at least two distinct RTS and a second gene of interest are chromosomally-integrated within the NL1 locus; and (c) at least two distinct RTS and a third gene of interest are chromosomally-integrated within the NL2 locus.
  • the present disclosure provides a method for producing a recombinant protein producer cell comprising (a) providing a cell that comprises at least at least four distinct RTS and a gene encoding a site-specific recombinase, wherein at least two distinct RTS are chromosomally-integrated within the NL1 locus and at least two distinct RTS are chromosomally-integrated within the NL2 locus; (b) transfecting the cell of (a) with a first vector comprising an exchangeable cassette encoding a first gene of interest and a second vector comprising an exchangeable cassette encoding a second gene of interest; (c) integrating the first exchangeable cassette within the NL1 locus and the second exchangeable cassette within the NL2 locus; and (d) selecting a recombinant protein producer cell comprising the first exchangeable cassette and the second exchangeable cassette integrated into the chromosome.
  • the first gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the DtE protein consists of a Fc-fusion protein, an enzyme, a membrane receptor, a bi-specific T-cell engager (BITE®), or a monoclonal antibody.
  • the monoclonal antibody is a bi-specific monoclonal antibody or a tri-specific monoclonal antibody.
  • the first gene of interest is located between two of the RTS.
  • the second gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein. In some embodiments, the second gene of interest is located between two of the RTS.
  • the present disclosure provides a method for producing a recombinant protein producer cell comprising (a) providing a cell that comprises at least four distinct RTS and a gene encoding a site-specific recombinase, wherein at least two distinct RTS are chromosomally-integrated within the Fer1L4 locus, and at least two distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus; (b) transfecting the cell of (a) with a first vector comprising an exchangeable cassette encoding a first gene of interest and a second vector comprising an exchangeable cassette encoding a second gene of interest; (c) integrating the first exchangeable cassette within the Fer1L4 locus and the second exchangeable cassette within the NL1 locus or the NL2 locus; and (d) selecting a recombinant protein producer cell comprising the first exchangeable cassette and the second exchangeable cassette integrated into the chromosome.
  • the first gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the DtE protein consists of a Fc-fusion protein, an enzyme, a membrane receptor, a bi-specific T-cell engager (BITE®), or a monoclonal antibody.
  • the monoclonal antibody is a bi-specific monoclonal antibody or a tri-specific monoclonal antibody.
  • the first gene of interest is located between two of the RTS.
  • the second gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein. In some embodiments, the second gene of interest is located between two of the RTS.
  • the present disclosure provides a method for producing a recombinant protein producer cell comprising (a) providing a cell that comprises at least at least six distinct RTS and a gene encoding a site-specific recombinase, wherein at least two distinct RTS are chromosomally-integrated within the Fer1L4 locus, and at least two distinct RTS are chromosomally-integrated within the NL1 locus, and at least two distinct RTS are chromosomally-integrated within the NL2 locus; (b) transfecting the cell of (a) with a first vector comprising an exchangeable cassette encoding a first gene of interest, a second vector comprising an exchangeable cassette encoding a second gene of interest, and a third vector comprising an exchangeable cassette encoding a third gene of interest; (c) integrating the first exchangeable cassette within the Fer1L4 locus, the second exchangeable cassette within the NL1 locus, and the third exchangeable cassette within the
  • FIG. 1 is a schematic for a method of recombinase-mediated cassette-exchange (RMCE) using a multisite site-specific integration (SSI) host cell line with 2 independently addressable landing pads.
  • the number of landing pads is restricted by the availability of suitable loci, incompatible Frt recombinase sites (e.g. wild-type Frt (F), Frt F5 (F5), Frt F14 (F14) and Frt F15 (F15)) and selection markers (e.g. green fluorescent protein (GFP) or Red Fluorescent Protein (RFP)) compatible with such a multiplexing approach.
  • F wild-type Frt
  • Frt F5 Frt F5
  • Frt F14 Frt F14
  • Frt F15 Frt F15
  • selection markers e.g. green fluorescent protein (GFP) or Red Fluorescent Protein (RFP)
  • FIG. 2A shows a schematic representation of the landing pad arrangement in a CHOK1SV GS-KOTM (glutamine synthetase knockout) multisite SSI host which contains two different illustrative landing pads (Landing Pad A and Landing Pad B). More details on the chromosomally-integration of these landing pads in single and multisite GS-CHOK1SVTM SSI hosts is given in Table 10.
  • Landing Pad A contains a hygromycin phosphotransferase (Hpt)—enhanced green fluorescent protein (eGFP) fusion gene (Hpt-eGFP).
  • Hpt-eGFP fusion gene expression is under the control of a SV40 early promoter and has an SV40 polyA sequence.
  • Distinct and different Frt sites are located between the SV40 promoter and Hpt-eGFP gene (F5) and 5′ of the SV40 poly A sequence (F) for RMCE.
  • Landing Pad B contains a puromycin N-acetyltransferase-DsRed (PAC-DsRed) fusion gene under the control of a SV40 promoter and includes a SV40 polyA sequence.
  • PAC-DsRed puromycin N-acetyltransferase-DsRed
  • a different pair of Frt sites, distinct from those in Landing Pad A, are located between the SV40 early promoter and PAC-DsRed gene (F14) and 5′ of the SV40 polyA sequence (F15).
  • Targeting vectors designed for RMCE in SSI single and multisite hosts contain a positive selection marker (e.g. GS) arranged immediately to the 3′ of an Frt site compatible with the destination landing pad ( FIG. 2B ). The remainder of the vector contains transcription units for the GOI (e.g. mAb) followed by an Frt site compatible to the second Frt site in the landing pad.
  • Targeting vector DNA FIG. 3A and FIG. 5 is co-transfected with a vector expressing FlpE recombinase (Takata et al., Genes to Cells 16: 7 (2011) ( FIG.
  • Transfected cells are incubated for 24 hours and selection pressure is then applied (e.g. removal of glutamine from culture medium).
  • Selection pressure is then applied (e.g. removal of glutamine from culture medium).
  • Successful RMCE is marked by the loss of the Hpt-eGFP gene and replacement with the positive selection marker gene ( FIG. 2C ).
  • Cells appear dark under the fluorescent microscope or with flow cytometer analysis. Non-exchanged cells which recover in positive selection are easily removed by fluorescence-aided cell sorting.
  • FIG. 3 shows the pMF25 targeting vector ( FIG. 3A ) and pMF4 recombinase expression vector ( FIG. 3B ), used for creating DsRed-producing cell lines in the CHOK1SV GS-KOTM SSI hosts.
  • pMF25 FIG. 3A
  • hCMV major intermediate early gene 1
  • Intron A hCMV Intron
  • Glutamine synthetase cDNA (GS) and SV40 Intron A (SV40 Inton) are arranged immediately to the 3′ of Frt F5 and successful RMCE transcription in driven by SV40E promoter located in the landing pad.
  • pMF4 FIG. 3B contains a transcription unit incorporating the FlpE gene (Takata et al., Genes to Cells 16: 7 (2011) driven by the promoter of the hCMV major intermediate early gene 1 (hCMV) and its first intron (Intron A (hCMV Intron)) and the flanking exons encoding the 5′ UTR are denoted as Ex1 and Ex2.
  • polyadenylation sequence and ⁇ -lactamase are indicated as pA and bla, respectively.
  • FIG. 4 shows the data from flow cytometry analysis of CHOK1SV GS-KOTM SSI host clones (7878, 8086, 8096, 9113, 9116 and 9115) prior to and following co-transfection with pMF25 and FlpE recombinase encoding vector.
  • Green and yellow fluorescence was measured for the 7878 and 8086 (single site, Fer1L4 Landing Pad A), 8086 and 9113 (single site, NL1 Landing Pad A) and 9116 and 9115 (single site, NL2 Landing Pad A) pre- and post-RMCE (11 days of selection in glutamine-free medium) using a Millipore Guava flow cytometer.
  • eGFP fluorescence was detected in the green channel and DsRed was detected in the yellow channel.
  • FIG. 5 shows the targeting vector for creating mAb producing cell lines in the CHOK1SV GS-KOTM derived SSI hosts.
  • Vectors pMF26 (A), pMF27 (B) and pMF28 (C) contain transcription units incorporating rituximab, cB72.3 and H31K5 antibody genes (respectively), flanked by mutant (F5) and wild-type (F) Frt sites.
  • Transcription of heavy (HC) and light chain (LC) genes are driven by the promoter of the hCMV major intermediate early gene 1 (hCMV) and its first intron, Intron A (hCMV Intron) and the flanking exons encoding the 5′ UTR are denoted as Ex1 and Ex2.
  • Glutamine synthetase cDNA (GS) and SV40 Intron-polyA (indicated as pA in the FIG. 5 ) sequences are arranged immediately to the 3′ of Frt F5 to select for on-target integration following RMCE.
  • ⁇ -lactamase is indicated as and bla, respectively.
  • FIG. 6 shows secreted Rituximab, cB72.3 and H31K5 mAb concentrations CHOK1SV GS-KOTM SSI pools grown in batch culture.
  • CHOK1SV GS-KOTM SSI host clone 11434 single site, Fer1L4 Landing Pad A
  • pMF26 Rhuximab
  • pMF27 cB72.3
  • pMF28 H31K5
  • Replicate n>3
  • CHOK1SV GS-KOTM SSI pools were cultured in 8-day batch culture.
  • Secreted rituximab (A), cB72.3 (B) and H31K5 (C) mAb (mg/L) were determined by Protein A HPLC.
  • FIG. 7 shows the data from flow cytometry analysis of multisite SSI hosts prior to RMCE.
  • Green and yellow fluorescence was measured for the CHOK1SV GS-KOTM host (Host), 11434 (single site, Fer1L4 Landing Pad A), DsRed random integration control and six CHOK1SV GS-KOTM multisite host clones (12151, 12152, 12606, 12607, 12608 and 12609) (multisite sister clones with Landing Pad A in the Fer1L4 loci and Landing Pad B in the NL1 loci) using a Millipore Guava flow cytometer. eGFP fluorescence was detected in the green channel and DsRed was detected in the yellow channel.
  • FIG. 8 is a schematic of the vectors pCM9 and pCM11 targeted to Fer1L4 and NL1 landing pads in the CHOK1SV GS-KO SSI host. Vectors are as follows: FIG. 8A : pCM9 (E2 crimson expression vector targeting Fer1L4 loci). FIG. 8B : pCM11 (E2 crimson expression vector targeting NL1 loci).
  • FIG. 9 shows flow cytometry analysis of CHOK1SV GS-KO SSI derived cells targeted with pCM9 and pCM11.
  • Multi CHOK1SV GS-KO SSI clone 12151 (Contains Landing Pad A ( FIG. 2A ) in Fer1L4 loci and Landing Pad B ( FIG. 2A ) in NL1 loci) was transfected with either pCM9 (E2 crimson expression vector targeting Fer1L4 loci) and pCM11 (E2 crimson expression vector targeting NL1 loci).
  • FIG. 10 is a schematic of the Rituximab expression vectors pMF26, pCM38, pCM22 and pAR5 targeted to either Fer1L4 ( FIG. 2 , Landing Pad A), NL1 ( FIG. 2 , Landing Pad B) or both.
  • Vectors were as follows: pMF26 ( FIG. 10A : Rituximab 1 ⁇ LC, 1 ⁇ HC expression vector targeting Fer1L4 loci), pCM38 ( FIG. 10B : Rituximab 2 ⁇ LC, 2 ⁇ HC expression vector targeting Fer1L4 loci), pCM22 ( FIG. 10C : empty expression vector targeting the NL1 loci) and pAR5 ( FIG. 10D : Rituximab 1 ⁇ LC, 1 ⁇ HC expression vector targeting NL1 loci).
  • FIG. 11 shows cell specific Rituximab production rates (qmAb) of RMCE pools targeted with pMF26, pCM38, pCM22 and pAR5 following an 8-day batch culture.
  • FIG. 12 is a schematic of the Cergutuzumab amunaleukin (CEA-IL2v) expression vectors pAB2, pAB5, pCM46 and pAR2, targeted to either Fer1L4 ( FIG. 2 , Landing Pad A), NL1 ( FIG. 2 , Landing Pad B) or both.
  • Vectors were as follows: pAB2 (CEA-IL2v LC, HC and HC-IL2 expression vector targeting Fer1L4 loci), pAB5 (CEA-IL2v LC and HC-IL2 expression vector targeting Fer1L4 loci), pCM46 (empty expression vector targeting NL1 loci) and pAR2 (CEA-IL2v HC expression vector targeting NL1 loci).
  • FIG. 13 is a schematic of SDS-page analysis of RMCE pools expressing Cergutuzumab amunaleukin (CEA-IL2v) following transfection with pAB2, pAB5, pCM46 and pAR2 ( FIG. 12 ).
  • A Image of a 10% Bis-Tris protein gel generated under non-reduced conditions with lanes 1-9 labelled. Lane 1: Seeblue Plus2 Pre-stained protein standard. Lane 2: Analysis of mock (empty) pool generated by transfecting an empty version of pAB2. Lanes 2-5: Control pools generated with vector expressing only two of three the chain required to generate CEA-IL2v used to assign folding intermediates.
  • Lane 6 analysis of a pools with CEA-IL2v LC, HC and HC-IL2 (pAB2) in Fer1L4 locus.
  • Lane 7 analysis of a pools with CEA-IL2v LC and HC-IL2 in Fer1L4 locus and empty vector (pCM46) in NL1.
  • Lanes 8 and 9 CEA-IL2v LC and HC-IL2 in Fer1L4 locus and CEA-IL2v HC (pAR2) in NL1 locus.
  • FIG. 14 is a schematic of the Entanercept, Ancillary or destabilized GFP (dsGFP with miR target sequence in 3′UTR) gene vectors targeted to either Fer1L4 ( FIG. 2 , Landing Pad A), NL1 ( FIG. 2 , Landing Pad B) or both.
  • Vectors were as follows: pTC1 ( FIGS. 14A , B and C: Entanercept expression vector targeting Fer1L4 loci), pCM22 ( FIG. 14A : Empty vector targeting NL1), pCM39 ( FIG. 14B : Mus musculus SCD1 (mSCD1) expression vector targeting NL1), pCM40 ( FIG.
  • FIG. 14B Mus musculus SCD1 codon optimized for Cricetulus griseus (ccmSCD1) expression vector targeting NL1), pCM41 ( FIG. 14B : Homo sapiens SCD1 (hSCD1), pCM42 ( FIG. 14B : Cricetulus griseus SREBF1 (ccSREBF1) expression vector targeting NL1), pCM43 ( FIG. 14C : dsGFP_6n CPEB2A expression vector targeting NL1), pCM44 ( FIG. 14C : dsGFP_6n CPEB2B expression vector targeting NL1) and pCM45 ( FIG. 14C : dsGFP_6n SRP ⁇ expression vector targeting NL1). See Table 11 and Table 12 for details of ancillary genes and sponge sequences.
  • the term “about” is used to indicate that a value includes the inherent variation of error for the method/device being employed to determine the value, or the variation that exists among the study subjects. Typically the term is meant to encompass approximately or less than 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19% or 20% variability depending on the situation.
  • the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, un-recited, elements or method steps. It is contemplated that any embodiment discussed in this specification can be implemented with respect to any method, system, host cells, expression vectors, and/or composition of the invention. Furthermore, compositions, systems, cells, and/or vectors of the invention can be used to achieve any of the methods as described herein.
  • the present disclosure provides a mammalian cell comprising at least two distinct RTS wherein the RTS are chromosomally-integrated within the NL1 locus or the NL2 locus.
  • the term “mammalian cell” includes cells from any member of the order Mammalia, such as, for example, human cells, mouse cells, rat cells, monkey cells, hamster cells, and the like.
  • the mammalian cell is a mouse cell, a human cell, a Chinese hamster ovary (CHO) cell, a CHO-K1 cell, a CHO-DXB11 cell, a CHO-DG44 cell, a CHOK1SVTM cell including all variants (e.g.
  • CHOK1SVTM POTELLIGENT® Lonza, Slough, UK
  • CHOK1SV GS-KOTM glutamine synthetase knockout
  • nucleic acid means a polymeric compound comprising covalently linked nucleotides.
  • nucleic acid includes polyribonucleic acid (RNA) and polydeoxyribonucleic acid (DNA), both of which may be single- or double-stranded.
  • DNA includes, but is not limited to, complimentary DNA (cDNA), genomic DNA, plasmid or vector DNA, and synthetic DNA.
  • RNA includes, but is not limited to, mRNA, tRNA, rRNA, snRNA, microRNA, miRNA, or MIRNA.
  • amino acid refers to a compound containing both a carboxyl (—COOH) and amino (—NH 2 ) group. “Amino acid” refers to both natural and unnatural, i.e., synthetic, amino acids.
  • Natural amino acids include alanine (Ala; A); arginine (Arg, R); asparagine (Asn; N); aspartic acid (Asp; D); cysteine (Cys; C); glutamine (Gln; Q); glutamic acid (Glu; E); glycine (Gly; G); histidine (His; H); isoleucine (Ile; I); leucine (Leu; L); lysine (Lys; K); methionine (Met; M); phenylalanine (Phe; F); proline (Pro; P); serine (Ser; S); threonine (Thr; T); tryptophan (Trp; W); tyrosine (Tyr; Y); and valine (Val; V).
  • peptide refers to a polymeric form of amino acids of any length, which can include coded and non-coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones.
  • chain and polypeptide “chain” are used interchangeably herein and refer to a polymeric form of amino acids of a single peptide backbone.
  • recombinant when used in reference to a nucleic acid molecule, peptide, polypeptide, or protein means of, or resulting from, a new combination of genetic material that is not known to exist in nature.
  • a recombinant molecule can be produced by any of the well-known techniques available in the field of recombinant technology, including, but not limited to, polymerase chain reaction (PCR), gene cutting (e.g., using restriction endonucleases), and solid state synthesis of nucleic acid molecules, peptides, or proteins.
  • PCR polymerase chain reaction
  • gene cutting e.g., using restriction endonucleases
  • solid state synthesis of nucleic acid molecules, peptides, or proteins solid state synthesis of nucleic acid molecules, peptides, or proteins.
  • “recombinant” refers to a viral vector or virus that is not known to exist in nature, e.g.
  • “recombinant” refers to a cell or host cell that is not known to exist in nature, e.g. a cell or host cell that has one or more mutations, nucleic acid insertions, or heterologous genes in the cell or host cell.
  • isolated polypeptide, protein, peptide, or nucleic acid is a molecule that has been removed from its natural environment. It is also to be understood that “isolated” polypeptides, proteins, peptides, or nucleic acids may be formulated with excipients such as diluents or adjuvants and still be considered isolated.
  • sequence identity or “% identity” in the context of nucleic acid sequences or amino acid sequences refers to the percentage of residues in the compared sequences that are the same when the sequences are aligned over a specified comparison window.
  • a comparison window can be a segment of at least 10 to over 1000 residues in which the sequences can be aligned and compared.
  • Methods of alignment for determination of sequence identity are well-known in the art can be performed using publicly available databases such as BLAST (blast.ncbi.nlm.nih.gov/Blast.cgi.).
  • polypeptides or nucleic acid molecules have at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, at least about 99% or about 100% sequence identity with a reference polypeptide or nucleic acid molecule, respectively (or a fragment of the reference polypeptide or nucleic acid molecule).
  • polypeptides or nucleic acid molecules have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99% or 100% sequence identity with a reference polypeptide or nucleic acid molecule, respectively (or a fragment of the reference polypeptide or nucleic acid molecule). In some embodiments, polypeptides or nucleic acid molecules have about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% sequence identity with a reference polypeptide or nucleic acid molecule, respectively.
  • a “gene” refers to an assembly of nucleotides that encode a polypeptide, and includes cDNA and genomic DNA nucleic acid molecules. “Gene” also refers to a nucleic acid fragment that can act as a regulatory sequence preceding (5′ non-coding sequences) and following (3′ non-coding sequences) the coding sequence. In some embodiments, genes are integrated in the host cell genome with multiple copies. In some embodiments, genes are integrated in the host cell genome at predefined copy numbers.
  • the term “regulatory element” refers to a genetic element which controls some aspect of the expression of nucleic acid sequences.
  • promoter refers to a DNA regulatory region/sequence capable of binding RNA polymerase and involved in initiating transcription of a downstream coding or non-coding sequence.
  • the promoter sequence includes the transcription initiation site and extends upstream to include the minimum number of bases or elements necessary to initiate transcription at levels detectable above background.
  • the promoter sequence includes a transcription initiation site, as well as protein binding domains responsible for the binding of RNA polymerase.
  • Eukaryotic promoters will often, but not always, contain “TATA” boxes and “CAT” boxes.
  • Various promoters including inducible promoters, may be used to drive the gene expression, e.g., in the host cell or vectors of the present disclosure.
  • the promoter is not a leaky promoter, i.e., the promoter is not constitutively expressing any one of the gene products as described herein.
  • heterologous promoter refers to such a regulatory element which is derived from a different species than the gene to which it is operably linked.
  • the heterologous promoter is derived from a prokaryotic system.
  • the heterologous promoter is derived from a eukaryotic system.
  • the disclosure provides for a cell in which one or more heterologous promoters are chromosomally-integrated into the host cell genome.
  • the terms “in operable combination,” “in operable order,” and “operably linked” refer to the linkage of nucleic acid sequences in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced. The term also refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced.
  • a gene of interest is operably linked to a promoter, wherein the gene of interest is chromosomally-integrated into the host cell.
  • the gene of interest is operably linked to a heterologous promoter; where in the gene of interest is chromosomally-integrated into the host cell.
  • an ancillary gene is operably linked to a promoter, wherein the ancillary gene is chromosomally-integrated into the host cell genome.
  • the ancillary gene is operably linked to a heterologous promoter; where in the ancillary gene is chromosomally-integrated into the host cell genome.
  • a gene encoding a DtE protein is operably linked to a promoter, wherein the gene encoding a DtE protein is chromosomally-integrated into the host cell genome.
  • the gene encoding a DtE protein is operably linked to a heterologous promoter, where in the gene encoding a DtE protein is chromosomally-integrated into the host cell genome.
  • a recombinase gene is operably linked to a promoter, wherein the recombinase gene is chromosomally-integrated into the host cell.
  • the recombinase gene is operably linked to a promoter, where in the recombinase gene is not integrated into the host cell genome.
  • a recombinase gene is operably linked to a heterologous promoter, wherein the recombinase gene is not chromosomally-integrated into the host cell genome. In some embodiments, the recombinase gene is operably linked to a heterologous promoter, wherein the recombinase gene is not chromosomally-integrated into the host cell genome.
  • regulatory elements operably link gene expression to the presence of an exogenously supplied ligand.
  • a gene of interest is operably linked to a promoter, wherein the promoter operably links gene expression to the presence of an exogenously supplied ligand and wherein the gene of interest is chromosomally-integrated into the host cell genome.
  • an ancillary gene is operably linked to a promoter, wherein the promoter operably links gene expression to the presence of an exogenously supplied ligand and wherein the ancillary gene is chromosomally-integrated into the host cell.
  • a gene encoding a DtE protein is operably linked to a promoter, wherein the promoter operably links gene expression to the presence of an exogenously supplied ligand and wherein the gene encoding a DtE protein is chromosomally-integrated into the host cell genome.
  • a recombinase gene is operably linked to a promoter, wherein the promoter operably links gene expression to the presence of an exogenously supplied ligand and wherein the recombinase gene is chromosomally-integrated into the host cell genome.
  • a recombinase gene is operably linked to a promoter, wherein the promoter operably links gene expression to the presence of an exogenously supplied ligand and wherein the recombinase gene is not chromosomally-integrated into the host cell genome.
  • chromosomally-integrated refers to the stable incorporation of a nucleic acid sequence into the chromosome of a host cell, e.g. a mammalian cell. i.e., a nucleic acid sequence that is chromosomally-integrated into the genomic DNA (gDNA) of a host cell, e.g. a mammalian cell.
  • a nucleic acid sequence that is chromosomally-integrated is stable.
  • a nucleic acid sequence that is chromosomally-integrated is not located on a plasmid or a vector.
  • a nucleic acid sequence that is chromosomally-integrated is not excised.
  • chromosomal integration is mediated by the clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR associated protein (Cas) gene editing system (CRISPR/CAS).
  • CRISPR clustered regularly interspaced short palindromic repeats
  • Cas CRISPR associated protein
  • the term “chromosomal locus” refers to defined location of nucleic acids on the chromosome of the cell that may comprise at least one gene.
  • the chromosomal locus comprises about 500 base pairs to about 100,000 base pairs, about 5,000 base pairs to about 75,000 base pairs, about 5,000 base pairs to about 60,000 base pairs, about 20,000 base pairs to about 50,000 base pairs, about 30,000 base pairs to about 50,000 base pairs, or about 45,000 base pairs to about 49,000 base pairs.
  • the chromosomal locus extends up to about 100 base pairs, about 250 base pairs, about 500 base pairs, about 750 base pairs, or about 1000 base pairs to the 5′ or the 3′ end of the defined nucleic acid sequence.
  • the chromosomal locus comprises an endogenous nucleic acid sequence. In some embodiments, the chromosomal locus comprises an exogenous nucleotide sequence having been integrated into the chromosome using methods known to one of the art of molecular biology. In some embodiments, the chromosomal locus comprises a nucleotide sequence in the genome of a host cell which provides for a strong and stable production of a heterologous protein encoded by a gene of interest integrated within the chromosomal locus. In some embodiments, the chromosomal locus comprises a nucleotide sequence in the genome of a host cell which provides for a strong and stable viral gene expression.
  • the chromosomal locus comprises a nucleotide sequence in the genome of a host cell which provides efficient site-specific integration.
  • the chromosomal locus comprises the NL1 locus, the NL2 locus, the NL3 locus, the NL4 locus, the NL5 locus, or the NL6 locus as described in Table 1.
  • the disclosure is directed to a mammalian cell comprising at least two distinct RTS wherein two RTS are chromosomally-integrated within the NL1 locus, the NL2 locus, the NL3 locus, the NL4 locus, the NL5 locus, or the NL6 locus as described in Table 1.
  • the disclosure is directed to a mammalian cell comprising at least two distinct RTS wherein two RTS are chromosomally-integrated within the NL1 locus or the NL2 locus as described in Table 1.
  • the term “integrated within the NL1 locus” or “integrated within the NL2 locus” will include integration into any part of the locus, and it not limited to just the indicated genomic coordinates.
  • the term “integrated within the NL1 locus” or “integrated within the NL2 locus” would also include corresponding loci in corresponding organisms.
  • the term “integrated within the NL1 locus” or “integrated within the NL2 locus” will include integration within about 50,000 bp, within about 40,000 bp, within about 30,000 bp, within 20,000 bp or within 10,000 bp of the indicated genomic coordinates.
  • the disclosure is directed to a mammalian cell comprising at least two distinct RTS wherein two RTS are chromosomally-integrated into a chromosomal locus selected from Fer1L4 (see e.g. U.S. patent application Ser. No. 14/409,283), ROSA26, HGPRT, DHFR, COSMC, LDHA, or MGAT1.
  • the chromosomal locus comprises the first intron of MID1 on the X chromosome.
  • the chromosomal locus comprises an enhanced expression and stability region (EESYRs see, e.g. U.S. Pat. No. 7,771,997).
  • EESYRs see, e.g. U.S. Pat. No. 7,771,997.
  • at least a portion of a gene in the native chromosomal locus is deleted.
  • a “vector” or “expression vector” is a replicon, such as a plasmid, phage, virus, or cosmid, to which another DNA segment may be attached to bring about the replication and/or expression of the attached DNA segment in a cell.
  • “Vector” includes episomal (e.g., plasmids) and non episomal vectors. In some embodiments of the present disclosure the vector is an episomal vector, which is removed/lost from a population of cells after a number of cellular generations, e.g., by asymmetric partitioning.
  • the term “vector” includes both viral and non-viral means for introducing a nucleic acid molecule into a cell in vitro, in vivo, or ex vivo.
  • the term vector may include synthetic vectors. Vectors may be introduced into the desired host cells by well-known methods, including, but not limited to, transfection, transduction, cell fusion, and lipofection. Vectors can comprise various regulatory elements including promoters
  • the term “exchangeable cassette” or “cassette” is a type of mobile genetic element that contains a gene and a recombination site.
  • the exchangeable cassette comprises at least two RTS.
  • the exchangeable cassette comprises a reporter gene or a selection gene.
  • a cassette is exchanged through recombinase-mediated cassette-exchange (RMCE).
  • site-specific integration is used to introduce one or more genes into a host cell chromosome. See, e.g., Bode et al., Biol. Chem. 381:801-813 (2000), Kolb, Cloning and Stem Cells 4:381-392 (2002) and Coates et al., Trends in Biotech. 23:407-419 (2005), each of which is incorporated by reference.
  • site-specific integration refers to integration of a nucleic acid sequence into a chromosome at a specific site.
  • site-specific integration can also mean “site-specific recombination.”
  • site-specific recombination refers to the rearrangement of two DNA partner molecules by specific enzymes performing recombination at their cognate pairs of sequences or target sites. Site-specific recombination, in contrast to homologous recombination, requires no DNA homology between partner DNA molecules, is RecA-independent, and does not involve DNA replication at any stage.
  • site-specific recombination uses a site-specific recombinase system to achieve site-specific integration of nucleic acids in host cells, e.g. mammalian cells.
  • a recombinase system typically consists of three elements: two specific DNA sequences (recombination target sites) and a specific enzyme (recombinase).
  • the recombinase catalyzes a recombination reaction between the specific recombination sites.
  • a recombinase enzyme, or recombinase is an enzyme that catalyzes recombination in site-specific recombination.
  • the recombinase used for site-specific recombination is derived from a non-mammalian system.
  • the recombinase is derived from bacteria, bacteriophage, or yeast.
  • a nucleic acid sequence encoding a recombinase is integrated into the host cell, e.g. mammalian cell. In some embodiments, a nucleic acid sequence encoding a recombinase is delivered to the host cell by methods known to molecular biology. In some embodiments, a recombinase polypeptide sequence can be delivered to the cell directly.
  • the recombinase is a Cre recombinase, a FLP recombinase, a Dre recombinase, a KD recombinase, a B2B3 recombinase, a Hin recombinase, a Tre recombinase, a ⁇ integrase, a HK022 integrase, a HP1 integrase, a ⁇ resolvase/invertase, a ParA resolvase/invertase, a Tn3 resolvase/invertase, a Gin resolvase/invertase, a ⁇ C31 integrase, a BxB1 integrase, a R4 integrase or another functional recombinase enzyme.
  • a recombinase as described herein is a FLP recombinase.
  • a FLP recombinase is a protein which catalyzes a site-specific recombination reaction that is involved in amplifying the copy number of the 2 plasmid of Saccharomyces cerevisiae during DNA replication.
  • the FLP recombinase of the present disclosure is derived from species of the genus Saccharomyces .
  • the FLP recombinase is derived from Saccharomyces cerevisiae .
  • the FPL recombinase is derived from a strain of Saccharomyces cerevisiae .
  • the FLP recombinase is a thermostable, mutant FLP recombinase.
  • the FLP recombinase is FLP1 or FLPe.
  • the nucleic acid sequence encoding the FLP recombinase comprises human optimized codons.
  • the recombinase is a Cre recombinase.
  • Cre causes recombination
  • Int family of recombinases Argonal recombinases
  • Cre has been shown to perform efficient recombination of lox sites (locus of X-ing over) not only in bacteria but also in eukaryotic cells (Sauer (1987) Mol. Cell. Biol. 7:2087; Sauer and Henderson (1988) Proc. Natl Acad. Sci. 85:5166).
  • the Cre recombinase as described and used herein is derived from bacteriophage.
  • the Cre recombinase is derived from P1 bacteriophage.
  • the terms “site-specific integration site,” “recombination target site,” “RTS,” and “site-specific recombinase target site” refer to a short, e.g. less than about 60 base pairs, nucleic acid site or sequence which is recognized by a site-specific recombinase and which become the crossover regions during the site-specific recombination event.
  • the recombination target site is less than about 60 base pairs, less than about 55 base pairs, less than about 50 base pairs, less than about 45 base pairs, less than about 40 base pairs, less than about 35 base pairs, or less than about 30 base pairs.
  • the recombination target site is about 30 to about 60 base pairs, about 30 to about 55 base pairs, about 32 to about 52 base pairs, about 34 to about 44 base pairs, about 32 base pairs, about 34 base pairs, or about 52 base pairs.
  • site-specific recombinase target sites include, but are not limited to, lox sites, rox sites, frt sites, att sites and dif sites.
  • recombination target sites are nucleic acids having substantially the same sequence as set forth in SEQ ID NOs.: 1-30.
  • the RTS is a lox site selected from Table 2.
  • lox site refers to a nucleotide sequence at which a Cre recombinase can catalyze a site-specific recombination.
  • a variety of non-identical lox sites are known to the art. The sequences of the various lox sites are similar in that they all contain identical 13-base pair inverted repeats flanking an 8-base pair asymmetric core region in which the recombination occurs. It is the asymmetric core region that is responsible for the directionality of the site and for the variation among the different lox sites.
  • loxP the sequence found in the P1 genome
  • loxB the sequence found in the P1 genome
  • loxL the sequence found in the E. coli chromosome
  • loxP 511 the sequence found in the P1 genome
  • lox ⁇ 86 the sequence found in the P1 genome
  • loxC 2 the sequence found in the E. coli chromosome
  • loxP 3 the sequence found in the E. coli chromosome
  • a lox recombination target site is a nucleic acid having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the sequences found in Table 2.
  • the RTS is a lox site selected from lox ⁇ 86, lox ⁇ 117, loxC2, loxP 2, loxP 3 and loxP 23.
  • the RTS is a Frt site selected from Table 3.
  • the term “Frt site” refers to a nucleotide sequence at which the product of the FLP gene of the yeast 2 ⁇ m plasmid, FLP recombinase, can catalyze a site-specific recombination.
  • a variety of non-identical Frt sites are known to the art. The sequences of the various Frt sites are similar in that they all contain identical 13-base pair inverted repeats flanking an 8-base pair asymmetric core region in which the recombination occurs. It is the asymmetric core region that is responsible for the directionality of the site and for the variation among the different Frt sites.
  • the Frt recombination target site is a nucleic acid having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the sequences found in Table 3.
  • the RTS is a rox site selected from Table 4.
  • rox site refers to a nucleotide sequence at which a Dre recombinase can catalyze a site-specific recombination.
  • roxR a nucleotide sequence at which a Dre recombinase can catalyze a site-specific recombination.
  • roxR a nucleotide sequence at which a Dre recombinase can catalyze a site-specific recombination.
  • roxR a variety of non-identical rox sites are known to the art. Illustrative (non-limiting) examples of these include roxR and roxF.
  • a rox recombination target site is a nucleic acid having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the sequences found in Table 4.
  • the RTS is an att site selected from Table 5.
  • att site refers to a nucleotide sequence at which a ⁇ integrase or ⁇ C31 integrase, can catalyze a site-specific recombination.
  • a variety of non-identical aat sites are known to the art. Illustrative (non-limiting) examples of these include attP, attB, proB, trpC, galT, thrA, and rrnB.
  • an att recombination target site is a nucleic acid having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the sequences found in Table 5.
  • distinct recombination target sites refers to non-identical or hetero-specific recombination target sites. For example, several variant Frt sites exist, but recombination can usually occur only between two identical Frt sites.
  • distinct recombination target sites refer to non-identical recombination target sites from the same recombination system (e.g. LoxP and LoxR).
  • distinct recombination target sites refer to non-identical recombination target sites from different recombination systems (e.g. LoxP and Frt).
  • distinct recombination target sites refer to a combination of recombination target sites from the same recombination system and recombination target sites from different recombination systems (e.g. LoxP, LoxR, Frt, and Frt1).
  • the cell comprises two RTS. In some embodiments, the cell comprises four RTS. In some embodiments, the cell comprises six RTS. In some embodiments, at least one RTS is selected from SEQ ID Nos.: 1-30. In some embodiments, the cell comprises six RTS. In some embodiments, at least one RTS is selected from SEQ ID Nos.: 1-6. In some embodiments, the cell comprises six RTS. In some embodiments, at least one RTS is selected from SEQ ID Nos.: 7-21. In some embodiments, the cell comprises six RTS. In some embodiments, at least one RTS is selected from SEQ ID Nos.: 22-23.
  • At least one RTS is selected from SEQ ID Nos.: 28-30. In some embodiments, the cell comprises six RTS. In some embodiments, at least one RTS is selected from SEQ ID Nos.: 24-27. In some embodiments, the cell comprising at least four distinct RTS includes the following RTS: LoxP 511, Frt F5, Frt, and LoxP. In some embodiments, the cell comprising at least four distinct RTS includes the following RTS: Frt F14 and Frt F15. In some embodiments, the cell comprising at least four distinct RTS includes the following RTS: LoxP 511, Frt F5, Frt, LoxP, Frt F14 and Frt F15.
  • the cell comprising at least four distinct RTS can include the following RTS: Frt 1, Frt 2, Frt 3, Frt 4. In some embodiments, the cell comprising at least four distinct RTS includes the following RTS: Frt m5, Frt wt, Frt m14, Frt m15, Frt m7 and Frt m6. In some embodiments, the cell comprising six distinct RTS can include the following RTS: Frt 1, Frt 2, Frt 3, Frt 4, Frt 5, Frt 6. In some embodiments, the cell comprising at least four distinct RTS can include the following RTS: Frt m5, Frt, Frt m14, Frt m15.
  • the term “landing pad” refers to a nucleic acid sequence comprising a first recombination target site chromosomally-integrated into a host cell.
  • a landing site comprises two or more recombination target sites chromosomally-integrated into a host cell.
  • the cell comprises 1, 2, 3, 4, 5, 6, 7, or 8 landing pads.
  • the cell comprises 1, 2, or 3 landing pads.
  • the cell comprises 4 landing pads.
  • landing pads are integrated at up to 1, 2, 3, 4, 5, 6, 7, or 8 distinct chromosomal loci.
  • landing pads are integrated at up to 1, 2, or 3 distinct chromosomal loci.
  • landing pads are integrated at 4 distinct chromosomal loci.
  • the disclosure describes how by expressing various proteins, e.g. DtE proteins, from various loci, it is possible to achieve the desired expression of the protein.
  • DtE protein is expressed from the NL1 locus, e.g., on the CHO chromosome.
  • the DtE protein is expressed from the NL2 locus, e.g., on the CHO chromosome.
  • the cell comprises two distinct RTS. In some embodiments, the two distinct RTS are chromosomally-integrated within the NL1 locus. In some embodiments, the two distinct RTS are chromosomally-integrated within the NL2 locus. In some embodiments, the cell comprises four distinct RTS. In some embodiments, the four distinct RTS are chromosomally-integrated on the same locus. In some embodiments, two distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus, and two distinct RTS are chromosomally-integrated on a separate locus. In some embodiments, the separate locus is the Fer1L4 locus.
  • two distinct RTS are chromosomally-integrated within the NL1 locus, and two distinct RTS are chromosomally-integrated within the NL2 locus.
  • the cell comprises six distinct RTS.
  • at least four distinct RTS are chromosomally-integrated on the same locus.
  • at least two distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus, and at least two distinct RTS are chromosomally-integrated on a separate locus.
  • the separate locus is the Fer1L4 locus.
  • At least two distinct RTS are chromosomally-integrated within the NL1 locus, and at least two distinct RTS are chromosomally-integrated within the NL2 locus.
  • at least one of the RTS is an frt site, a lox site, a rox site, or an att site.
  • at least one of the RTS is selected from among SEQ ID NOS.: 1-30.
  • the cell is a mouse cell, a human cell, a Chinese hamster ovary (CHO) cell, a CHO-K1 cell, a CHO-DXB11 cell, a CHO-DG44 cell, a CHOK1SVTM cell including all variants (e.g. CHOK1SVTM POTELLIGENT®, Lonza, Slough, UK), a CHOK1SV GS-KOTM (glutamine synthetase knockout) cell including all variants, a HEK293 cell including adherent and suspension-adapted variants, a HeLa cell, or a HT1080 cell.
  • CHO Chinese hamster ovary
  • the mammalian cell comprises at least two distinct RTS, wherein the RTS are chromosomally-integrated within the NL1 locus, the NL2 locus or the Fer1L4 locus. In some embodiments, the mammalian cell comprises at least four distinct RTS wherein two distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus and wherein two distinct RTS are chromosomally-integrated within a separate locus. In some embodiments, the separate locus is the Fer1L4 locus.
  • the mammalian cell comprises at least four distinct RTS, wherein two distinct RTS are chromosomally-integrated within the NL1 locus and two distinct RTs are chromosomally-integrated within the NL2 locus. In some embodiments, the mammalian cell comprises at least four distinct RTS wherein four distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus.
  • the mammalian cell comprises at least six distinct RTS, where two distinct RTS are chromosomally-integrated within the NL1 locus, two distinct RTS are chromosomally-integrated within the NL2 locus, and two distinct RTS are chromosomally-integrated at a separate locus.
  • the separate locus is the Fer1L4 locus.
  • the mammalian cell comprises at least six distinct RTS, wherein six distinct RTS are integrated within the NL1 locus or the NL2 locus.
  • the mammalian cell comprises at least six distinct RTS, wherein four distinct RTS are integrated at the NL1 locus or the NL2 locus and wherein two distinct RTS are integrated at a separate locus.
  • the second locus is the Fer1L4 locus.
  • the cell comprises a first gene of interest, wherein the first gene of interest is chromosomally-integrated.
  • heterologous gene As referred to herein, the term “gene of interest” or “GOI” is used to describe a heterologous gene.
  • the term “heterologous gene” or “HG” as it relates to nucleic acid sequences such as a coding sequence or a control sequence denotes a nucleic acid sequence, e.g. a gene, that is not normally joined together, and/or are not normally associated with a particular cell.
  • a heterologous gene is a construct where the coding sequence itself is not found in nature (e.g., synthetic sequences having codons different from the native gene). Allelic variation or naturally occurring mutational events do not give rise to heterologous DNA, as used herein.
  • the gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene or a combination thereof.
  • a “reporter gene” is a gene whose expression confers a phenotype upon a cell that can be easily identified and measured.
  • the reporter gene comprises a fluorescent protein gene.
  • the reporter gene comprises a selection gene.
  • selection gene refers to the use of a gene which encodes an enzymatic activity that confers the ability to grow in medium lacking what would otherwise be an essential nutrient; in addition, a selection gene may confer resistance to an antibiotic or drug upon the cell in which the selection gene is expressed.
  • a selection gene may be used to confer a particular phenotype upon a host cell. When a host cell must express a selection gene to grow in selective medium, the gene is said to be a positive selection gene. Selection gene can also be used to select against host cells containing a particular gene; selection genes used in this manner are referred to as negative selection genes.
  • the terms “gene of therapeutic interest” refers to any functionally relevant nucleotide sequence.
  • the gene of therapeutic interest of the present disclosure can comprise any desired gene that encodes a protein the expression of which is desired the preparation of a therapeutic recombinant protein.
  • suitable genes of therapeutic interest include monoclonal antibodies, bi-specific monoclonal antibodies, or antibody drug conjugates [include blood clotting factors, well expressed mAbs where protein expression is limited at transcription, hormones such as EPO, immune-fusion proteins (Fc fusions), tri-specific mAbs].
  • the terms “ancillary gene” or “helper gene” are used interchangeable to refer to a first gene that aids in the expression of a second gene or that aids in the stabilization, folding, or post translational modification of the product of the second gene or that creates a cellular environment that promotes the production of the product of the second gene.
  • the second gene is a gene encoding a DtE protein.
  • the ancillary gene encodes RNA.
  • the ancillary gene encodes an mRNA, a tRNA, or a miRNA.
  • the ancillary gene encodes a transcription factor, a chaperone, a chaperonin, a synthetase, an oxidase, a reductase, a glycotransferase, a protease, a kinase, a phosphatase, an acetyl transferase, a lipase, or an alkylase.
  • the first gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a well expressed therapeutic protein at a desired copy number.
  • the gene encoding a well expressed therapeutic protein is at a copy number of 2 copies, of 3 copies, of 4 copies, of 5 copies, of 6 copies, of 7 copies, of 8 copies, of 9 copies, or of 10 copies.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the term a “difficult to express protein” refers to a protein for which production is difficult.
  • product of the DtE protein can be difficult because protein expression must be highly regulated.
  • the DtE protein is difficult to recover from the host cell.
  • the DtE protein is a protein that is prone to mis-folding.
  • the DtE protein is a protein that is prone to clipping.
  • the DtE protein is a protein that is prone to degradation.
  • the DtE protein is a protein that is prone to aggregation.
  • the DtE protein is a protein that is poorly soluble.
  • a DtE protein is a membrane bound protein. In some embodiments, the DtE protein is difficult to purify. In some embodiments, a DtE protein is cytotoxic. In some embodiments, the DtE protein comprises multiple polypeptide chains, e.g. 2, 3 or 4 polypeptide chains. In some embodiments, the multiple polypeptide chains of the DtE protein form a homo-oligomer to produce the DtE protein. In some embodiments, the multiple polypeptide chains of the DtE protein form a hetero-oligomer to produce the DtE protein. In some embodiments, the homo-oligomer or the hetero-oligomer is formed through covalent interactions, non-covalent interactions, or a combination thereof.
  • the DtE protein comprises a protein for which the expression of an ancillary gene is required to produce the DtE protein. In some embodiments, the DtE protein is a protein for which a post-translational modification is required to produce the DtE protein. In some embodiments, the DtE protein is a protein for which expression using standard techniques known to one of the art of molecular biology, results in a product protein with variable characteristics. In some embodiments, the DtE protein is a fusion protein.
  • the disclosure describes how expressing DtE proteins from NL1 and/or NL2 locus that DtE proteins can be obtained in more than 2 g/L protein production titers.
  • the expression of a DtE protein at the NL1 locus yields more than 2 g/L of the DtE protein.
  • the expression of a DtE protein at the NL2 locus yields more than 2 g/L of the DtE protein.
  • the DtE protein is a protein for which expression using standard techniques known to one of the art of molecular biology, results in a product protein titer less than 2 g/L.
  • the DtE protein is a monoclonal antibody. In some embodiments, the DtE protein is a bi-specific monoclonal antibody. In some embodiments, the DtE protein is a tri-specific monoclonal antibody. In some embodiments, the DtE protein is an Fc-fusion protein. As referred to herein, the term “Fc-fusion protein” refers to a fusion protein wherein the Fc domain of an immunoglobulin is operably linked to a second peptide. In some embodiments, the DtE protein is an enzyme. In some embodiments, the DtE protein is a membrane receptor. In some embodiments, the DtE protein is a bi-specific T-cell engager (BITE® Micromet AG, Kunststoff, Germany).
  • the DtE protein is selected from the group consisting of an Fc-fusion protein, an enzyme, a membrane receptor, or a monoclonal antibody.
  • the monoclonal antibody is a bi-specific monoclonal antibody or a tri-specific monoclonal antibody.
  • the DtE protein is encoded on one or more genes of interest.
  • the first gene of interest is located between two of the RTS.
  • the term “located between two of the RTS” refers to a gene located between two of the RTS, i.e., with one of the RTS located 5′ of the gene and a different RTS located 3′ of the gene.
  • the RTS are located directly adjacent to the gene located between them.
  • the RTS are located at a defined distance from the gene located between them.
  • the RTS are directional sequences.
  • the RTS 5′ and 3′ of the gene located between them are directly oriented (i.e. they are oriented in the same direction).
  • the RTS 5′ and 3′ of the gene located between them are inversely oriented (i.e. they are oriented in opposite directions).
  • the first gene of interest is located within the NL1 locus.
  • the cell comprises a second gene of interest, wherein the second gene of interest is chromosomally-integrated.
  • the second gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the DtE protein is selected from the group consisting of a Fc-fusion protein, an enzyme, a membrane receptor, a bi-specific T-cell engager (BITE®), or a monoclonal antibody.
  • the second gene of interest is located between two of the RTS. In some embodiments, the second gene of interest is located within the NL1 locus or the NL2 locus. In some embodiments, the first gene of interest is located within the NL1 locus, and the second gene of interest is located within the NL2 locus. In some embodiments, the cell comprises a third gene of interest, wherein the third gene of interest is chromosomally-integrated. In some embodiments, the third gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof. In some embodiments, the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the third gene of interest is located between two of the RTS. In some embodiments, the third gene of interest is located within the NL1 locus or the NL2 locus. In some embodiments, the third gene of interest is located with a locus distinct from the NL1 locus and the NL2 locus. In some embodiments, the first gene of interest, the second gene of interest, and the third gene of interest are within three separate loci. In some embodiments, at least one of the first genes of interest, the second gene of interest, and the third gene of interest is within the NL1 locus, and at least one of the first gene of interest, the second gene of interest, and the third gene of interest is within the NL2 locus.
  • the cell comprises a site-specific recombinase gene.
  • the site-specific recombinase gene is chromosomally-integrated.
  • the present disclosure provides a mammalian cell comprising (a) at least four distinct RTS, wherein the cell comprises at least two distinct RTS are chromosomally-integrated within the NL1 locus or NL2 locus; (b) a first gene of interest is integrated between the at least two RTS of (a), wherein the first gene of interest comprises a reporter gene, a gene encoding a DtE protein, an ancillary gene or a combination thereof, and (c) a second gene of interest is integrated within a second chromosomal locus distinct from the locus of (a), wherein the second gene of interest comprises a reporter gene, a gene encoding a DtE protein, an ancillary gene or a combination thereof.
  • the present disclosure provides a mammalian cell comprising (a) at least four distinct RTS, wherein the cell comprises at least two distinct RTS are chromosomally-integrated within the Fer1L4 locus; (b) at least two distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus; (c) a first gene of interest is chromosomally-integrated within the Fer1L4 locus, wherein the first gene of interest comprises a reporter gene, a gene encoding a DtE protein, an ancillary gene or a combination thereof; and (d) a second gene of interest is chromosomally-integrated within the within the NL1 locus or NL2 locus of (b), wherein the second gene of interest comprises a reporter gene, a gene encoding a DtE protein, an ancillary gene or a combination thereof.
  • the present disclosure provides a mammalian cell comprising at least six distinct RTS, wherein the cell comprises (a) at least two distinct RTS and a first gene of interest are chromosomally-integrated within the Fer1L4 locus; (b) at least two distinct RTS and a second gene of interest are chromosomally-integrated within the NL1 locus; and (c) at least two distinct RTS and a third gene of interest are chromosomally-integrated within the NL2 locus.
  • the present disclosure provides a method for producing a recombinant protein producer cell comprising (a) providing a cell that comprises at least four distinct RTS and a gene encoding a site-specific recombinase, wherein at least two distinct RTS are chromosomally-integrated within the NL1 locus and at least two distinct RTS are chromosomally-integrated within the NL2 locus; (b) transfecting the cell of (a) with a first vector comprising an exchangeable cassette encoding a first gene of interest and a second vector comprising an exchangeable cassette encoding a second gene of interest; (c) integrating the first exchangeable cassette within the NL1 locus and the second exchangeable cassette within the NL2 locus; and (d) selecting a recombinant protein producer cell comprising the first exchangeable cassette and the second exchangeable cassette integrated into the chromosome.
  • Transfection means the introduction of an exogenous nucleic acid molecule, including a vector, into a cell.
  • a “transfected” cell comprises an exogenous nucleic acid molecule inside the cell and a “transformed” cell is one in which the exogenous nucleic acid molecule within the cell induces a phenotypic change in the cell.
  • the transfected nucleic acid molecule can be integrated into the host cell's genomic DNA and/or can be maintained by the cell, temporarily or for a prolonged period of time, extra-chromosomally.
  • Host cells or organisms that express exogenous nucleic acid molecules or fragments are referred to as “recombinant,” “transformed,” or “transgenic” organisms.
  • transfection techniques are generally known in the art. See, e.g., Graham et al., Virology, 52:456 (1973); Sambrook et al., Molecular Cloning, a laboratory manual, Cold Spring Harbor Laboratories, New York (1989); Davis et al., Basic Methods in Molecular Biology, Elsevier (1986); and Chu et al., Gene 13:197 (1981).
  • Such techniques can be used to introduce one or more exogenous DNA moieties into suitable host cells.
  • an RTS of an exchangeable cassette matching an RTS of the cell refers to the RTS of the cassette having a sequence substantially identical to the RTS of the cell.
  • the exchangeable cassette contains a sequence substantially identical to one or two of the RTS chromosomally-integrated into the host cell genome.
  • the term integrating refers to the integration, e.g. insertion, of the exchangeable cassette into the chromosome.
  • integration is mediated by a site-specific recombinase.
  • the inventors find that the use of SSI eliminates the need to clone cells from those transfected, as the cells are homogenous in their genetic composition.
  • the term “selecting” refers to identifying cells containing a chromosomally-integrated marker. In some embodiments, selection is through the detection of the presence of a marker using methods known to those skilled in the art. In some embodiments, selection is through the detection of the absence of a marker using methods known to those skilled in the art.
  • the first gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the DtE protein consists of a Fc-fusion protein, an enzyme, a membrane receptor, a bi-specific T-cell engager (BITE®), or a monoclonal antibody.
  • the monoclonal antibody is a bi-specific monoclonal antibody or a tri-specific monoclonal antibody.
  • the first gene of interest is located between two of the RTS.
  • the second gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein. In some embodiments, the second gene of interest is located between two of the RTS.
  • the present disclosure provides a method for producing a recombinant protein producer cell comprising (a) providing a cell that comprises at least at least four distinct RTS and a gene encoding a site-specific recombinase, wherein at least two distinct RTS are chromosomally-integrated within the Fer1L4 locus, and at least two distinct RTS are chromosomally-integrated within the NL1 locus or the NL2 locus; (b) transfecting the cell of (a) with a first vector comprising an exchangeable cassette encoding a first gene of interest and a second vector comprising an exchangeable cassette encoding a second gene of interest; (c) integrating the first exchangeable cassette within the Fer1L4 locus and the second exchangeable cassette within the NL1 locus or the NL2 locus; and (d) selecting a recombinant protein producer cell comprising the first exchangeable cassette and the second exchangeable cassette integrated into the chromosome.
  • the first gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein.
  • the DtE protein consists of a Fc-fusion protein, an enzyme, a membrane receptor, a bi-specific T-cell engager (BITE®), or a monoclonal antibody.
  • the monoclonal antibody is a bi-specific monoclonal antibody or a tri-specific monoclonal antibody.
  • the first gene of interest is located between two of the RTS.
  • the second gene of interest comprises a reporter gene, a selection gene, a gene of therapeutic interest, an ancillary gene, or a combination thereof.
  • the gene of therapeutic interest comprises a gene encoding a DtE protein. In some embodiments, the second gene of interest is located between two of the RTS.
  • the present disclosure provides a method for producing a recombinant protein producer cell comprising (a) providing a cell that comprises at least at least six distinct RTS and a gene encoding a site-specific recombinase, wherein at least two distinct RTS are chromosomally-integrated within the Fer1L4 locus, and at least two distinct RTS are chromosomally-integrated within the NL1 locus, and at least two distinct RTS are chromosomally-integrated within the NL2 locus; (b) transfecting the cell of (a) with a first vector comprising an exchangeable cassette encoding a first gene of interest, a second vector comprising an exchangeable cassette encoding a second gene of interest, and a third vector comprising an exchangeable cassette encoding a third gene of interest; (c) integrating the first exchangeable cassette within the Fer1L4 locus, the second exchangeable cassette within the NL1 locus, and the third exchangeable cassette within the
  • the inventors have found that the use of SSI to prepare rP expression cells ensures the pool of rP expression cells is homogenous in its genetic makeup. In some embodiments, the inventors have found that the use of SSI to prepare rP expression cells ensures the pool of rP expression cells is homogenous in its efficiency. In some embodiments, the inventors have found that the use of SSI to prepare rP expression cells ensures the pool of producer cells is homogenous in the ratio of a first helper gene to a second helper gene. In some embodiments, the inventors have found that the use of SSI to prepare rP expression cells ensures the pool of producer cells is homogenous in the ratio of helper genes to genes of therapeutic interest. In some embodiments, the inventors have found that the use of SSI to prepare rP expression cells ensures more consistent rP product quality.
  • the cell lines described herein can be cultured using any suitable device, facility and methods described herein.
  • the devices, facilities and methods are suitable for culturing suspension cells or anchorage-dependent (adherent) cells and are suitable for production operations configured for production of pharmaceutical and biopharmaceutical products-such as polypeptide products, nucleic acid products (for example DNA or RNA), or mammalian or microbial cells and/or viruses such as those used in cellular and/or viral and microbiota therapies.
  • the cells express or produce a product, such as a recombinant therapeutic or diagnostic product.
  • a product such as a recombinant therapeutic or diagnostic product.
  • products produced by cells include, but are not limited to, antibody molecules (e.g., monoclonal antibodies, bispecific antibodies), antibody mimetics (polypeptide molecules that bind specifically to antigens but that are not structurally related to antibodies such as e.g.
  • DARPins affibodies, adnectins, or IgNARs
  • fusion proteins e.g., Fc fusion proteins, chimeric cytokines
  • other recombinant proteins e.g., glycosylated proteins, enzymes, hormones
  • viral therapeutics e.g., anti-cancer oncolytic viruses, viral vectors for gene therapy and viral immunotherapy
  • cell therapeutics e.g., pluripotent stem cells, mesenchymal stem cells and adult stem cells
  • vaccines or lipid-encapsulated particles e.g., exosomes, virus-like particles
  • RNA such as e.g. siRNA
  • DNA such as e.g. plasmid DNA
  • antibiotics or amino acids antibiotics or amino acids.
  • the devices, facilities and methods can be used for producing biosimilars.
  • devices, facilities and methods allow for the production of eukaryotic cells, e.g., mammalian cells or lower eukaryotic cells such as for example yeast cells or filamentous fungi cells, or prokaryotic cells such as Gram-positive or Gram-negative cells and/or products of the eukaryotic or prokaryotic cells, e.g., proteins, peptides, antibiotics, amino acids, nucleic acids (such as DNA or RNA), synthesized by the eukaryotic cells in a large-scale manner.
  • prokaryotic cells e.g., proteins, peptides, antibiotics, amino acids, nucleic acids (such as DNA or RNA), synthesized by the eukaryotic cells in a large-scale manner.
  • proteins, peptides, antibiotics, amino acids, nucleic acids (such as DNA or RNA), synthesized by the eukaryotic cells in a large-scale manner e.g., proteins, peptides, antibiotics, amino acids
  • the devices, facilities, and methods can include any suitable reactor or bioreactor including but not limited to stirred tank, airlift, fiber, microfiber, hollow fiber, ceramic matrix, fluidized bed, fixed bed, and/or spouted bed bioreactors.
  • reactor or “bioreactor” can include a fermenter or fermentation unit, or any other reaction vessel and the term “reactor” is used interchangeably with “fermenter.”
  • fermenter or fermentation refers to both microbial and mammalian cultures.
  • an example bioreactor unit can perform one or more, or all, of the following: feeding of nutrients and/or carbon sources, injection of suitable gas (e.g., oxygen), inlet and outlet flow of fermentation or cell culture medium, separation of gas and liquid phases, maintenance of temperature, maintenance of oxygen and CO2 levels, maintenance of pH level, agitation (e.g., stirring), and/or cleaning/sterilizing.
  • suitable gas e.g., oxygen
  • Example reactor units such as a fermentation unit, may contain multiple reactors within the unit, for example the unit can have 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, or 100, or more bioreactors in each unit and/or a facility may contain multiple units having a single or multiple reactors within the facility.
  • the bioreactor can be suitable for batch, semi fed-batch, fed-batch, perfusion, and/or a continuous fermentation processes. Any suitable reactor diameter can be used. In embodiments, the bioreactor can have a volume between about 100 mL and about 50,000 L.
  • Non-limiting examples include a volume of 100 mL, 250 mL, 500 mL, 750 mL, 1 liter, 2 liters, 3 liters, 4 liters, 5 liters, 6 liters, 7 liters, 8 liters, 9 liters, 10 liters, 15 liters, 20 liters, 25 liters, 30 liters, 40 liters, 50 liters, 60 liters, 70 liters, 80 liters, 90 liters, 100 liters, 150 liters, 200 liters, 250 liters, 300 liters, 350 liters, 400 liters, 450 liters, 500 liters, 550 liters, 600 liters, 650 liters, 700 liters, 750 liters, 800 liters, 850 liters, 900 liters, 950 liters, 1000 liters, 1500 liters, 2000 liters, 2500 liters, 3000 liters, 3
  • suitable reactors can be multi-use, single-use, disposable, or non-disposable and can be formed of any suitable material including metal alloys such as stainless steel (e.g., 316L or any other suitable stainless steel) and Inconel, plastics, and/or glass.
  • metal alloys such as stainless steel (e.g., 316L or any other suitable stainless steel) and Inconel, plastics, and/or glass.
  • the devices, facilities, and methods described herein can also include any suitable unit operation and/or equipment not otherwise mentioned, such as operations and/or equipment for separation, purification, and isolation of such products.
  • Any suitable facility and environment can be used, such as traditional stick-built facilities, modular, mobile and temporary facilities, or any other suitable construction, facility, and/or layout.
  • modular clean-rooms can be used.
  • the devices, systems, and methods described herein can be housed and/or performed in a single location or facility or alternatively be housed and/or performed at separate or multiple locations and/or facilities.
  • the cells are eukaryotic cells, e.g., mammalian cells.
  • the mammalian cells can be for example human or rodent or bovine cell lines or cell strains. Examples of such cells, cell lines or cell strains are e.g.
  • mouse myeloma e.g., NS0 or SP2/0 cell lines
  • Chinese hamster ovary (CHO) cell lines HT1080, H9, HepG2, MCF7, MDBK Jurkat, NIH3T3, PC12, BHK (baby hamster kidney cell), VERO, YB2/0, Y0, C127, L cell, COS, e.g., COS1 and COS7, QC1-3, HEK-293, VERO, PER.C6, HeLa, EB1, EB2, EB3, oncolytic or hybridoma-cell lines.
  • the mammalian cells are CHO-cell lines.
  • the cell is a CHO cell.
  • the cell is a CHO-K1 cell, a CHO-K1 SV cell, a DG44 CHO cell, a DUXB11 CHO cell, a CHOS, a CHO GS knock-out cell, a CHO FUT8 GS knock-out cell, a CHOZN, or a CHO-derived cell.
  • the CHO GS knock-out cell e.g., GSKO cell
  • the CHO FUT8 knockout cell is, for example, the CHOK1SVTM POTELLIGENT® (Lonza Biologics, Inc.).
  • Eukaryotic cells can also be avian cells, cell lines or cell strains, such as for example, EBX® cells, EB14, EB24, EB26, EB66, or EBvl3.
  • the eukaryotic cells are stem cells.
  • the stem cells can be, for example, pluripotent stem cells, including embryonic stem cells (ESCs), adult stem cells, induced pluripotent stem cells (iPSCs), tissue specific stem cells (e.g., hematopoietic stem cells) and mesenchymal stem cells (MSCs).
  • ESCs embryonic stem cells
  • iPSCs induced pluripotent stem cells
  • tissue specific stem cells e.g., hematopoietic stem cells
  • MSCs mesenchymal stem cells
  • the cell is a differentiated form of any of the cells described herein.
  • the cell is a cell derived from any primary cell in culture.
  • the cells are not derived from stem cells.
  • the cells are used in immunotherapies (e.g., lymphocytes) either extracted or isolated from individual patients or from established cell banks.
  • the cells can include genetically manipulated cells (i.e. CAR-T, etc.)
  • the cell is a hepatocyte such as a human hepatocyte, animal hepatocyte, or a non-parenchymal cell.
  • the cell can be a plateable metabolism qualified human hepatocyte, a plateable induction qualified human hepatocyte, plateable Qualyst Transporter CertifiedTM human hepatocyte, suspension qualified human hepatocyte (including 10-donor and 20-donor pooled hepatocytes), human hepatic Kupffer cells, human hepatic stellate cells, dog hepatocytes (including single and pooled Beagle hepatocytes), mouse hepatocytes (including CD-1 and C57BI/6 hepatocytes), rat hepatocytes (including Sprague-Dawley, Wistar Han, and Wistar hepatocytes), monkey hepatocytes (including Cynomolgus or Rhesus monkey hepatocytes), cat hepatocytes (including Domestic Shorthair hepatocytes), and
  • the eukaryotic cell is a lower eukaryotic cell such as e.g. a yeast cell (e.g., Pichia genus (e.g. Pichia pastoris, Pichia methanolica, Pichia kluyveri , and Pichia angusta ), Komagataella genus (e.g. Komagataella pastoris, Komagataella pseudopastoris or Komagataella phaffii ), Saccharomyces genus (e.g. Saccharomyces cerevisiae, Saccharomyces kluyveri, Saccharomyces uvarum ), Kluyveromyces genus (e.g.
  • a yeast cell e.g., Pichia genus (e.g. Pichia pastoris, Pichia methanolica, Pichia kluyveri , and Pichia angusta ), Komagataella genus (e.g. Komagat
  • Kluyveromyces lactis, Kluyveromyces marxianus the Candida genus (e.g. Candida utilis, Candida cacaoi, Candida boidinii ), the Geotrichum genus (e.g. Geotrichum fermentans ), Hansenula polymorpha, Yarrowia lipolytica , or Schizosaccharomyces pombe .
  • Candida genus e.g. Candida utilis, Candida cacaoi, Candida boidinii
  • Geotrichum genus e.g. Geotrichum fermentans
  • Hansenula polymorpha Yarrowia lipolytica
  • Schizosaccharomyces pombe e.g. Saccharin
  • Pichia pastoris examples are X33, GS115, KM71, KM71H; and CBS7435.
  • the eukaryotic cell is a fungal cell (e.g. Aspergillus (such as A. niger, A. fumigatus, A. orzyae, A. nidula ), Acremonium (such as A. thermophilum ), Chaetomium (such as C. thermophilum ), Chrysosporium (such as C. thermophile ), Cordyceps (such as C. militaris ), Corynascus, Ctenomyces, Fusarium (such as F. oxysporum ), Glomerella (such as G. graminicola ), Hypocrea (such as H. jecorina ), Magnaporthe (such as M.
  • Aspergillus such as A. niger, A. fumigatus, A. orzyae, A. nidula
  • Acremonium such as A. thermophilum
  • Chaetomium such as C. thermophilum
  • Chrysosporium such
  • orzyae Myceliophthora (such as M. thermophile ), Nectria (such as N. heamatococca ), Neurospora (such as N. crassa ), Penicillium, Sporotrichum (such as S. thermophile ), Thielavia (such as T. terrestris, T. heterothallica ), Trichoderma (such as T. reesei ), or Verticillium (such as V. dahlia )).
  • M. thermophile such as M. thermophile
  • Nectria such as N. heamatococca
  • Neurospora such as N. crassa
  • Penicillium such as S. thermophile
  • Thielavia such as T. terrestris, T. heterothallica
  • Trichoderma such as T. reesei
  • Verticillium such as V. dahlia
  • the eukaryotic cell is an insect cell (e.g., Sf9, MIMICTM Sf9, Sf21, HIGH FIVETM (BT1-TN-5B1-4), or BT1-Ea88 cells), an algae cell (e.g., of the genus Amphora, Bacillariophyceae, Dunaliella, Chlorella, Chlamydomonas, Cyanophyta (cyanobacteria), Nannochloropsis, Spirulina , or Ochromonas ), or a plant cell (e.g., cells from monocotyledonous plants (e.g., maize, rice, wheat, or Setaria ), or from a dicotyledonous plants (e.g., cassava, potato, soybean, tomato, tobacco, alfalfa, Physcomitrella patens or Arabidopsis ).
  • insect cell e.g., Sf9, MIMICTM Sf9, Sf21, HIGH FIVETM (BT1-
  • the cell is a bacterial or prokaryotic cell.
  • the prokaryotic cell is a Gram-positive cell such as Bacillus, Streptomyces Streptococcus, Staphylococcus or Lactobacillus.
  • Bacillus that can be used is, e.g. the B. subtilis, B. amyloliquefaciens, B. licheniformis, B. natto , or B. megaterium .
  • the cell is B. subtilis , such as B. subtilis 3NA and B. subtilis 168.
  • Bacillus is obtainable from, e.g., the Bacillus Genetic Stock Center, Biological Sciences 556, 484 West 12 th Avenue, Columbus Ohio 43210-1214.
  • the prokaryotic cell is a Gram-negative cell, such as Salmonella spp. or Escherichia coli , such as e.g., TG1, TG2, W3110, DH1, DHB4, DH5a, HMS 174, HMS174 (DE3), NM533, C600, HB101, JM109, MC4100, XL1-Blue and Origami, as well as those derived from E. coli B-strains, such as for example BL-21 or BL21 (DE3), all of which are commercially available.
  • Salmonella spp. or Escherichia coli such as e.g., TG1, TG2, W3110, DH1, DHB4, DH5a, HMS 174, HMS174 (DE3), NM533, C600, HB101, JM109, MC4100, XL1-Blue and Origami, as well as those derived from E. coli B-strains, such as for example
  • Suitable host cells are commercially available, for example, from culture collections such as the DSMZ (Deutsche Sammlung von Mikroorganismen and Zellkulturen GmbH, Braunschweig, Germany) or the American Type Culture Collection (ATCC).
  • the cells include other microbiota utilized as therapeutic agents. These include microbiota present in the human microbiome belonging to the phyla Firmicutes, Bacteroidetes, Proteobacteria, Verrumicrobia, actinobacteria, fusobacteria and cyanobacteria.
  • Microbiota can include both aerobic, strict anaerobic or facultative anaerobic and include cells or spores.
  • Therapeutic Microbiota can also include genetically manipulated organisms and vectors utilized in their modification.
  • microbiome-related therapeutic organisms can include: archaea, fungi and virus. See, e.g., The Human Microbiome Project Consortium. Nature 486, 207-214 (14 Jun. 2012); Weinstock, Nature, 489(7415): 250-256 (2012); Lloyd-Price, Genome Medicine 8:51 (2016).
  • the cultured cells are used to produce proteins e.g., antibodies, e.g., monoclonal antibodies, and/or recombinant proteins, for therapeutic use.
  • the cultured cells produce peptides, amino acids, fatty acids or other useful biochemical intermediates or metabolites.
  • molecules having a molecular weight of about 4000 Daltons to greater than about 140,000 Daltons can be produced.
  • these molecules can have a range of complexity and can include post-translational modifications including glycosylation.
  • the protein is, e.g., BOTOX, Myobloc, Neurobloc, Dysport (or other serotypes of botulinum neurotoxins), alglucosidase alpha, daptomycin, YH-16, choriogonadotropin alpha, filgrastim, cetrorelix, interleukin-2, aldesleukin, teceleulin, denileukin diftitox, interferon alpha-n3 (injection), interferon alpha-nl, DL-8234, interferon, Suntory (gamma-la), interferon gamma, thymosin alpha 1, tasonermin, DigiFab, ViperaTAb, EchiTAb, CroFab, nesiritide, abatacept, alefacept, Rebif, eptoterminalfa, teriparatide (osteoporosis), calcitonin injectable (bone disease), calcitonin (
  • the polypeptide is adalimumab (HUMIRA), infliximab (REMICADETM), rituximab (RITUXANTM/MABTHERATM) etanercept (ENBRELTM), bevacizumab (AVASTINTM), trastuzumab (HERCEPTINTM), pegrilgrastim (NEULASTATM), or any other suitable polypeptide including biosimilars and biobetters.
  • polypeptides are those listed below in Table 6 and in Table 1 of US2016/0097074.
  • One of skill in the art can appreciate that the disclosure of the present invention additional would encompass combinations of products and/or conjugates as described herein [(i.e. multi-proteins, modified proteins (conjugated to PEG, toxins, other active ingredients)].
  • the polypeptide is a hormone, blood clotting/coagulation factor, cytokine/growth factor, antibody molecule, fusion protein, protein vaccine, or peptide as shown in Table 7.
  • the protein is multispecific protein, e.g., a bispecific antibody as shown in Table 8.
  • GS-CHOK1SVTM clonal cell lines (Lonza, Basel, Switzerland) producing monoclonal antibody (mAb), constructed using random integration, were screened for those which met the following criteria: high qmAb (>1.25 pg/cell ⁇ h), stable productivity (>70 generations) and suitable growth (IVCC>1500 ⁇ 10 6 cells/mL ⁇ h, ⁇ ⁇ 0.03 h ⁇ 1 ).
  • MSX methionine sulfoximine
  • the lowest generation number ampoule for these twenty cell lines was cultured to mid-exponential culture phase for analysis of integrated vector copy number using qPCR (primer/probe sets for beta lactamase gene).
  • Five cell lines (964E7, 952C8, 2A6, E22 and E14) identified as meeting copy number criteria ( ⁇ 5 beta lactamase copies per genome) were sampled for flanking sequence identification.
  • Genomic DNA (gDNA) extracts were prepared with cells derived from the aforementioned 5 cell lines and were subjected to sequence capture analysis.
  • NimbleGen SeqCap Target Enrichment (Roche NimbleGen, Inc., Madison, USA) was completed on fragmented gDNA derived from recombinant cell lines using baits designed for regions within the glutamine synthase (GS) expression vector bearing mAb genes.
  • Target enriched pools were eluted and sequenced by Illumina MISEQ® (Next Gen Sequencing, Illumina, San Diego, Calif., USA).
  • WGRS whole genome re-sequencing
  • TLA targeted locus amplification
  • Bioinformatic analysis of sequencing reads was conducted. Capture sequencing data were mapped to genome and vector sequences. Split reads (half from genome, half from vector) were used to identify potential integration sites and the break point. In addition, read pairs that suggest vector insertion (one end on genome, one end on vector) were also identified to provide support evidence for the integration sites. A list of potential integration sites was identified from sequence capture data, and the sites were ranked by the supporting evidence. WGRS data were mapped to all potential integration sites to provide additional support evidence. Reads from WGRS that map across a break point, as well as read pairs that cover the break points were counted as positive supports.
  • left (L) and right (R) were used to refer to the locations of the genomic sequence (always 5′ to 3′ according to Cricetulus griseus scaffold and contig sequences).
  • the vector was inserted in the forward (F) or reverse direction (R).
  • Data relating to cell lines 964E7 and 952C8 were later by targeted sequencing with proximity ligation at Cergentis (Utrecht, NL) see, e.g. de Vree et al., Nat Biotechnol. 32:1019-25 (2014).
  • Integration sites were validated by PCR amplification across predicted genome: vector boundaries in all five cell lines. Table 9 summarizes these integration site findings, along-side the beta-lactamase gene copy number, productivity and growth data for these recombinant cell lines. A total of six sites (New Loci 1-6 (NL1-6)) were confirmed by PCR in the five cells lines, three which were confirmed by PCR across both genome vector boundaries (NL1, NL2 and NL4) and three which were confirmed at only one of the boundaries (NL3, NL5 and NL6).
  • New Loci 1-6 NL1-6
  • Loci NL1 and NL2 were progressed to SSI landing pad integration, as derivative cell lines 964E7 and 952C8 had similar integrated beta-lactamase copies to the other 3 cell lines, but achieved a higher specific productivity (47-48 pg/cell ⁇ day) suggesting these regions support higher recombinant gene expression.
  • the selection of loci from recombinant cell lines generated in CHOK1SVTM derivative hosts and using a GS selection marker ensured that loci are compatible with a GS expression system based system. These loci have been shown to support stable recombinant gene expression (qP: 23-48 pg/cells ⁇ day) without negatively affecting process important to growth (IVCC: 2039 to 6015 ⁇ 10 6 cells/h ⁇ mL).
  • Landing pads suitable for subsequent RMCE were integrated into the CHOK1SV GS-KOTM host cell line.
  • a landing pad (Landing Pad A: FIG. 2 ) was initially integrated separately into Fer1L4 (see, e.g., WO2013190032A1 and EP2711428A1) ( FIG. 4 : Clones 7878 and 8086, FIG. 7 : Clone 11434), NL1 ( FIG. 4 : Clones 8096 and 9113) and NL2 (Clones 9116 and 9115) loci.
  • Clone 11434 (landing pad in the Fer1L4 locus, Landing Pad A: FIG. 2A ), was selected for engineering of the second landing pad at NL1 (Landing Pad B: FIG. 2A ).
  • These 2-site hosts have a landing pad in Fer1L4 containing Hpt-eGFP fusion flanked by Frt F5 and wild-type Frt F RTS and a second landing pad in site 2 containing a PAC-DsRed fusion gene flanked by Frt F14 and Frt F15 RTS (landing pad in the NL1 loci, Landing Pad B: FIG. 2 ).
  • the positioning of the Frt site between the SV40E promoter and selection marker enables it to be used in subsequent rounds of RMCE.
  • Targeting vectors designed for RMCE in the CHOK1SV GS-KOTM single and multi-landing pad hosts contained the GS cDNA arranged immediately to the 3′ of a Frt site compatible with the destination landing pad ( FIG. 2B ). The remainder of the vector contained transcription units for the GOI (e.g., mAb) followed by a Frt site compatible to the second Frt site in the landing pad.
  • Targeting vector DNA FIG. 3A
  • FIG. 3B were co-transfected with a vector expressing FlpE recombinase ( FIG. 3B ) (at a plasmid molar ratio of 1:9, respectively).
  • Transfected cells were incubated for 24 hours in the presence of 6 mM glutamine to allow transient expression of the FlpE recombinase. This transfectant pool was then washed and incubated in medium lacking glutamine. The viable cell concentration and culture viability were monitored throughout selection.
  • Successful RMCE was marked by the loss of the Hpt-eGFP gene and replaced with GS gene in the targeting vector.
  • a no-FlpE control was included in all transfections (transfection of targeting vector DNA without pMF4) to confirm any recovery is the result of transient FlpE recombinase expression (RMCE). Upon successful RMCE cells appeared dark under fluorescent microscope or with flow cytometry analysis.
  • Targeting vector pMF25 ( FIG. 3A ) and recombinase vector pMF4 ( FIG. 3B ) were co-transfected into 6 CHOK1SV GS-KOTM ( FIG. 4 : Fer1L4 loci: 7878 and 8086, NL1: 8096 and 9113, NL2: 9116 and 9115) single landing pad SSI hosts and incubated in glutamine-free medium to select for cells which have completed RMCE.
  • CHOK1SV GS-KOTM SSI pools were analyzed by flow cytometry prior to RMCE and after 11 days in glutamine-free medium ( FIG.
  • the SSI host clone 11434 (landing pad in the Fer1L4 loci, Landing Pad A: FIG. 2 ) was then tested for the ability to produce therapeutic mAbs.
  • Vectors containing transcription units for rituximab, cB72.3 and H31K5 which target the Fer1L4 locus in clone 11434 were created (See FIG. 5 ).
  • CHOK1SV GS-KOTM pools were then constructed and cultured in batch suspension culture for 8 days. The concentration of secreted mAb at harvest was determined by Protein A HPLC at harvest (See FIG. 6 ). These data show very consistent expression between replicated pools and between different mAbs (250-300 mg/L).
  • the 6 multisite hosts described in Example 2 were analyzed by flow cytometry to confirm the ability of loci to support recombinant gene expression ( FIG. 7 ).
  • the landing pad integrated at the Fer1L4 loci ( FIG. 2 : Landing Pad A) contains the Hpt-eGFP gene and the landing pad integrated at NL1 loci ( FIG. 2 : Landing Pad B) contains the PAC-DsRed gene. eGFP fluorescence was detected in the green channel and DsRed was detected in the yellow channel.
  • the CHOK1SV GS-KOTM host (Host) was used as a negative control. Clone 11434 (landing pad in the Fer1L4 loci, Landing Pad A: FIG.
  • FIG. 8A contains E2 crimson expression cassette and targets landing pad A: Fer1L4) and pCM11 ( FIG. 8B , contains E2 crimson expression cassette and targets landing pad A: NL1) were transfected into multi-site CHOK1SV GS-KOTM SSI host 12151. Pools were incubated in glutamine-free medium to select for cells which have completed RMCE. The no-FlpE controls did not recover with RMCE pools. These CHOK1SV GS-KOTM SSI pools were analyzed by flow cytometry prior to RMCE and after 14 days in glutamine-free medium ( FIG. 8 ). As landing pad A in the CHOK1SV GS-KOTM SSI host (Landing Pad A, FIG.
  • Landing pad B in the CHOK1SV GS-KOTM SSI host contains the PAC-DsRed reporter however the DsRed signal was not detected on the flow cytometer ( FIG. 9 ).
  • pCM11 targeting vector contains E2 crimson reporter and therefore successful RMCE without the loss of Landing Pad A (containing Hpt-eGFP gene) was demonstrated by a change from +GFP, ⁇ RFP to +GFP, +RFP ( FIG. 9 ).
  • the CHOK1SVTM SSI multisite hosts (See FIG. 2 ) were tested for the expression of therapeutic proteins outlined in Table 10. Experiments are separated into three phases; Phase 1: Tests the application of multisite SSI to increase qP; Phase 2: Tests the capability of multisite SSI to express a three-gene bispecific mAb across the two landing pads; Phase 3: To express ancillary genes in one site in order to aid the expression of a DtE protein encoded in the other site.
  • FIGS. 10A pMF26 were transfected separately (in duplicate) into the GS-KO SSI host clone 12151 ( FIGS. 10A and B) and pools selected in glutamine-free medium (for a detailed transfection method see Example 2).
  • the neomycin phosphotransferase gene NEO
  • a version of pAR5 which lacked mAb genes was also created and referred to as pCM22 ( FIG. 10C ).
  • CHOK1SV GS-KOTM SSI pools transfected with pMF26 and selected in glutamine-free medium were then transfected (in duplicate) with either pAR5 ( FIG. 10D ) or pCM22 ( FIG. 10C ) (for a detailed transfection method see Example 2), incubated in the presence of pMF4 ( FIG. 3B ) for 24 hours and then selected in 400 ⁇ g/mL Geneticin (G418). Consistent with selection in glutamine free medium, No-FlpE controls cultured in the presence of G418 did not recover with RMCE pools. Following recovery from selection, the eight RMCE pools were sub-cultured and then progressed to an 8-day batch culture.
  • the viable cell concentration was determined at days 4, 6 and 8 using a Vicell cell counter and concentration of secreted mAb at harvest was determined by ForteBio Octet using Protein A sensors. Cell specific production rate of Rituximab (qmAb at harvest) was calculated (See FIG. 11 ). These data demonstrated that both scenarios are viable options for increasing cell specific mAb production rates from the CHOK1SV GS-KOTM SSI host.
  • next generation antibodies e.g. tetravalent bispecific antibodies
  • assembly of multiple heavy or light chains is a recurrent problem.
  • selection of an appropriate CHO clone expressing as many as four antibody chains in a stable and reproducible is beneficial.
  • product analytics utilizing ELISAs, RP-HPLC or CD-SDS during clone selection is often required.
  • the genes encoding a multi chain protein are driven with individual promoters and are spatially separated across at least two sites.
  • FIG. 12B CHOK1SV GS-KOTM SSI host was transfected with pAB5 ( FIG. 12B ) which contains expression units for cergutuzumab amunaleukin LC and HC-IL2 targeting landing pad A ( FIG. 2 ). Selection was in the absence of glutamine as described in Example 2. Subsequently we generated a targeting vector that contained a single cergutuzumab amunaleukin HC expression cassette ( FIG. 12C : pAR2) in addition to the neomycin phosphotransferase selection marker gene (NEO) which targeted landing pad B (Landing Pad B, FIG. 2A ).
  • NEO neomycin phosphotransferase selection marker gene
  • FIG. 12B A version of pAR2 which lacked mAb gene was also created and referred to as pCM46 ( FIG. 12B : pCM46).
  • CHOK1SV GS-KOTM SSI pools transfected with pAB5 and selected in glutamine-free medium were then transfected (in duplicate) with either pCM46 or pAR2 (for a detailed transfection method see Example 2) and selected in medium containing 400 ⁇ g/mL Geneticin (G418).
  • Control pools were also constructed: a ‘Mock’ transfection with an empty version of pAR2, and three pools, each which lacked one of the three genes encoding cergutuzumab amunaleukin (LC+HC, LC+HC-IL2 and HC+HC-IL2), to be used for identifying CEA-IL2v antibody species.
  • the RMCE pools were sub-cultured and then progressed to an 8-day batch culture. The viable cell concentration was determined at days 4, 6 and 8 using a Vicell cell counter. Cergutuzumab amunaleukin assembly species were determined by non-reduced analysis of supernatants on 10% Bis-Tris SDS PAGE gels ( FIG.
  • pCM39 to pCM45 contain expression units for ancillary genes under the control of the human cytomegalovirus major immediate-early (hCMV) promoter (with its first Intron A) and targets landing pad B ( FIG. 14 ) (Table 11 and 12).
  • hCMV human cytomegalovirus major immediate-early
  • Scd1 stearoyl-CoA desaturase-1
  • SREBF1 Sterol regulatory element-binding protein 1
  • a short-lived GFP (or dsGFP) designed with a c-terminus PEST sequence (SEQ ID NO: 38) from sequence gb:CQ871827 was constructed and 6 copies of the miR Target Sequence inserted into 3′UTR for CPEB2A (pCM43), CEPB2B (pCM44) and SRP ⁇ (pCM45).
  • These targeting vectors contain the neomycin phosphotransferase gene (NEO) and 24 hours following transfection (with pMF4) of each vector into duplicate pTC01 transfected pools, selection was achieved using medium supplemented with 400 ⁇ g/mL G418.
  • IVCC Integral of viable cell concentration
  • qP at harvest cell specific production rate
  • secreted etanercept concentration are presented (See FIG. 15 ).
  • mice SCD1 mouse SCD1
  • sponge vectors bearing CEPB2A dsGFP_6n CPEB2A
  • CPEB2B dsGFP_6n CPEB2B
  • SRP ⁇ dsGFP_6n SRP ⁇ miR binding site
  • CHOK1SV derived vector integration sites and landing pad locations were BLAT searched against the human genome (version: March 2006 (NCBI36/hg18)) using a stand-alone copy of the University of California Santa Cruz (UCSC) human genome data base. This identifies sequences of 95% (and greater) similarity, in at least 25 base pairs of CHOK1SV sequence. Regions of similarity were visualized in IGV viewer (Broad institute version 2.4). Crispr-Cas9 gRNA were design using an in house Crispr-Cas9 design tool. CHOK1SV and HEK293 loci are summarized in Table 1.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Mycology (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
US16/486,363 2017-02-17 2018-02-17 Multi-site specific integration cells for difficult to express proteins Pending US20200002727A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/486,363 US20200002727A1 (en) 2017-02-17 2018-02-17 Multi-site specific integration cells for difficult to express proteins

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762460420P 2017-02-17 2017-02-17
US16/486,363 US20200002727A1 (en) 2017-02-17 2018-02-17 Multi-site specific integration cells for difficult to express proteins
PCT/IB2018/000232 WO2018150269A1 (en) 2017-02-17 2018-02-17 Multi-site specific integration cells for difficult to express proteins

Publications (1)

Publication Number Publication Date
US20200002727A1 true US20200002727A1 (en) 2020-01-02

Family

ID=62002155

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/486,363 Pending US20200002727A1 (en) 2017-02-17 2018-02-17 Multi-site specific integration cells for difficult to express proteins

Country Status (8)

Country Link
US (1) US20200002727A1 (ko)
EP (1) EP3583121A1 (ko)
JP (2) JP7467119B2 (ko)
KR (1) KR102630357B1 (ko)
CN (1) CN111372946A (ko)
CA (1) CA3053712A1 (ko)
IL (1) IL268523A (ko)
WO (1) WO2018150269A1 (ko)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021084429A1 (en) 2019-11-01 2021-05-06 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2021165928A2 (en) 2020-02-23 2021-08-26 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2022090893A2 (en) 2020-10-27 2022-05-05 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2022137078A1 (en) 2020-12-23 2022-06-30 Pfizer Inc. E. coli fimh mutants and uses thereof
WO2023012627A1 (en) 2021-08-02 2023-02-09 Pfizer Inc. Improved expression vectors and uses thereof

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11202103111TA (en) * 2018-10-01 2021-04-29 Lonza Ag Ssi cells with predictable and stable transgene expression and methods of formation
EP3986925A1 (en) * 2019-06-19 2022-04-27 F. Hoffmann-La Roche AG Method for the generation of a trivalent antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization
US20220403398A1 (en) 2019-11-14 2022-12-22 Lonza Ltd Methods of cell selection
EP3901266A1 (en) * 2020-04-22 2021-10-27 LEK Pharmaceuticals d.d. Super-enhancers for recombinant gene expression in cho cells
JP2023525204A (ja) 2020-05-12 2023-06-15 ロンザ ヒューストン インコーポレイテッド アデノ随伴ウイルスを検出するための方法およびキット
KR102335242B1 (ko) * 2020-05-22 2021-12-02 인천대학교 산학협력단 Fer1L4 유전자에 부위-특이적 통합된 RMCE 랜딩 패드를 포함하는 CHO 세포
GB202019484D0 (en) * 2020-12-10 2021-01-27 Univ Edinburgh CHO cell modification
CN112920279B (zh) * 2021-03-09 2022-07-05 海南大学 一种海水提铀用抗生物污损型聚合肽水凝胶材料及其制备方法和应用

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5190871A (en) 1989-06-12 1993-03-02 Eli Lilly And Company Use of the site-specific integrating function of phage φC31
IT1258959B (it) 1992-06-09 1996-03-11 Impianto a moduli mobili per lo sviluppo e la produzione di prodotti biotecnologici su scala pilota
MXPA03010626A (es) 2001-05-30 2004-12-06 Chromos Molecular Systems Inc Plataformas a base de cromosomas.
GB0216648D0 (en) 2002-07-18 2002-08-28 Lonza Biologics Plc Method of expressing recombinant protein in CHO cells
MXPA06008724A (es) 2004-02-03 2007-02-16 Xcellerex Llc Sistema y metodo para elaboracion.
CN101001945B (zh) 2004-06-04 2012-02-08 艾克塞勒雷克斯公司 一次性生物反应器系统及方法
EP1815001B8 (en) * 2004-11-26 2011-03-23 Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Gene trap cassettes for random and targeted conditional gene inactivation
EP2004798A4 (en) 2005-12-05 2010-11-24 Ernest G Hope PREVALIDED, MODULAR GOODS MANUFACTURING PRACTICE CORRESPONDING ESTABLISHMENT
PT2137655E (pt) 2007-04-16 2012-09-14 Momenta Pharmaceuticals Inc Produtos de glicoproteína definidos e métodos relacionados
EP2150617B1 (en) 2007-06-04 2014-10-22 Regeneron Pharmaceuticals, Inc. Enhanced expression and stability regions
US8771635B2 (en) 2010-04-26 2014-07-08 Toyota Motor Engineering & Manufacturing North America, Inc. Hydrogen release from complex metal hydrides by solvation in ionic liquids
US10371394B2 (en) 2010-09-20 2019-08-06 Biologics Modular Llc Mobile, modular cleanroom facility
EP2447717B1 (en) * 2010-10-27 2013-09-18 Lonza Biologics plc. Rapid method for targeted cell (line) selection
US9388373B2 (en) 2011-03-08 2016-07-12 University Of Maryland Baltimore County Microscale bioprocessing system and method for protein manufacturing
US8980579B2 (en) * 2011-04-05 2015-03-17 The Scripps Research Institute Chromosomal landing pads and related uses
EP2527448A1 (en) * 2011-05-23 2012-11-28 Novozymes A/S Simultaneous site-specific integrations of multiple gene-copies in filamentous fungi
AR089793A1 (es) * 2012-01-27 2014-09-17 Du Pont Metodos y composiciones para generar locus de rasgos transgenicos complejos
EP2711428A1 (en) 2012-09-21 2014-03-26 Lonza Biologics plc. Site-specific integration
EP3030656B1 (en) 2013-08-06 2018-08-22 Lonza Biologics Plc. Means and methods for the generation of mammalian producer cells for the production of recombinant proteins
US10385352B2 (en) * 2015-03-09 2019-08-20 Novozymes A/S Methods of introducing multiple expression constructs into a eukaryotic cell

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
deVree (Nature Biotechnology, 32(10), 1019-1025 (Year: 2014) *
Lewis et al Nature Biotechnology, 31, 759-765 (Year: 2013) *
NCBI Blast sequence search of SEQ ID NO 32, page 1 (Year: 2023) *
Povey et al Journal of Biotechnology 184 84–93 (Year: 2014) *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021084429A1 (en) 2019-11-01 2021-05-06 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2021165928A2 (en) 2020-02-23 2021-08-26 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2022090893A2 (en) 2020-10-27 2022-05-05 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2022137078A1 (en) 2020-12-23 2022-06-30 Pfizer Inc. E. coli fimh mutants and uses thereof
WO2023012627A1 (en) 2021-08-02 2023-02-09 Pfizer Inc. Improved expression vectors and uses thereof

Also Published As

Publication number Publication date
CN111372946A (zh) 2020-07-03
KR20190129858A (ko) 2019-11-20
JP2023065343A (ja) 2023-05-12
EP3583121A1 (en) 2019-12-25
WO2018150269A1 (en) 2018-08-23
JP2020511954A (ja) 2020-04-23
KR102630357B1 (ko) 2024-01-30
WO2018150269A8 (en) 2019-03-21
JP7467119B2 (ja) 2024-04-15
IL268523A (en) 2019-09-26
CA3053712A1 (en) 2018-08-23

Similar Documents

Publication Publication Date Title
US20200002727A1 (en) Multi-site specific integration cells for difficult to express proteins
US11781116B2 (en) Mammalian cells for producing adeno-associated viruses
US20240132830A1 (en) Method and system for providing buffer solutions
CN108610398B (zh) 一段功能序列及在分泌蛋白表达中的应用
EP3741853B1 (en) Methods of cell selection and modifying cell metabolism
ES2921137T3 (es) Producción de proteínas regulada por fuente de carbono en una célula huésped recombinante
JP2023109812A (ja) 生物製剤の製造のためのユニバーサル自己調節性哺乳動物細胞株プラットフォーム
US20200332241A1 (en) Continuous Blade Impeller
US20220049275A1 (en) Ssi cells with predictable and stable transgene expression and methods of formation
US11965152B2 (en) Buffer formulation method and system
US11034721B2 (en) Method for the reduction of viral titer in pharmaceuticals
US20210324390A1 (en) Methods for improving production of biological products by reducing the level of endogenous protein
KR20230075436A (ko) 번역 인자를 과발현하는 숙주 세포
IL261411B1 (en) Improved fermentation process

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: PFIZER INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CASPERSON, GERALD FRIES;JONES, HEATHER LAURENCE;MOFFAT, MARK ALLEN;AND OTHERS;SIGNING DATES FROM 20200116 TO 20200117;REEL/FRAME:051845/0467

Owner name: LONZA LTD, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FEARY, MARC;YOUNG, ROBERT J.;SIGNING DATES FROM 20190820 TO 20190821;REEL/FRAME:051845/0423

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION